CN107922406B - CGRP receptor antagonists - Google Patents

CGRP receptor antagonists Download PDF

Info

Publication number
CN107922406B
CN107922406B CN201680043902.0A CN201680043902A CN107922406B CN 107922406 B CN107922406 B CN 107922406B CN 201680043902 A CN201680043902 A CN 201680043902A CN 107922406 B CN107922406 B CN 107922406B
Authority
CN
China
Prior art keywords
methyl
ethyl
pyrrolo
scheme
give
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201680043902.0A
Other languages
Chinese (zh)
Other versions
CN107922406A (en
Inventor
D·A·科茨
K·C·福特纳
R·D·约翰斯顿
S·M·麦西
J·K·迈尔斯
Q·史
M·G·西格尔
Original Assignee
Eli Lilly and Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly and Co filed Critical Eli Lilly and Co
Publication of CN107922406A publication Critical patent/CN107922406A/en
Application granted granted Critical
Publication of CN107922406B publication Critical patent/CN107922406B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C31/00Saturated compounds having hydroxy or O-metal groups bound to acyclic carbon atoms
    • C07C31/02Monohydroxylic acyclic alcohols
    • C07C31/04Methanol
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pain & Pain Management (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides compounds of formula II:

Description

CGRP receptor antagonists
The present invention relates to certain novel calcitonin gene-related peptide (CGRP) receptor antagonist compounds, pharmaceutical compositions comprising said compounds, methods of using said compounds for the prevention or treatment of physiological disorders such as migraine, and intermediates and processes for the synthesis of said compounds.
The present invention is in the field of prevention and treatment of migraine and other neurological diseases and disorders believed to be mediated by CGRP (see, e.g., s. benemei et al, Current Opinion in Pharmacology,9,9-14 (2009)). Migraine is a debilitating disease suffered by millions of people worldwide. Treatment options for migraine include triptans, such as sumatriptan and zolmitriptan. Unfortunately, the drugs currently used by approved patients do not always provide effective treatment, and these drugs may be associated with various untoward side effects such as dizziness, paresthesia, and chest discomfort. In addition, triptans have certain cardiovascular problems that make them contraindicated for use in patients with severe underlying cardiovascular disease or uncontrolled hypertension (see t.w. ho et al, The Lancet, 372, 2115-. Therefore, there is a significant unmet need in the prevention and treatment of migraine. There is a need for CGRP antagonists that provide more effective treatment or prevention of certain neurological diseases such as migraine.
U.S. patent No.4,960,785 discloses certain 2-oxo-indoline compounds as aldose reductase inhibitors for the control of certain chronic diabetic complications. In addition, T.Ooi et al, J Am. chem.Soc.,135(50),18706-18709(2013) disclose asymmetric substitutions on tetrasubstituted chiral carbons by catalytic ring-opening alkylation of racemic 2, 2-disubstituted aziridines with 3-substituted oxindoles.
The present invention provides certain novel compounds which are antagonists of CGRP receptors. In addition, the present invention provides certain novel compounds which are CGRP receptor antagonists with improved side effect profiles in the treatment or prevention of migraine.
Accordingly, the present invention provides a compound of formula II:
Figure GDA0002319866120000021
wherein
Y is CH or N;
z is CH or N;
provided that when Y is CH, Z is N, and when Y is N, Z is CH;
x is CH or N;
q is CH or N; and is
R is C1-C3 alkyl, C3-C5 cycloalkyl or CN;
or a pharmaceutically acceptable salt thereof.
The present invention also provides compounds of formula I:
Figure GDA0002319866120000022
or a pharmaceutically acceptable salt thereof.
The present invention also provides a method of preventing migraine in a patient comprising administering to a patient in need thereof an effective amount of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
The present invention also provides a method of treating migraine in a patient comprising administering to a patient in need thereof an effective amount of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof. The present invention also provides a method of antagonizing CGRP receptors in a patient comprising administering to a patient in need thereof an effective amount of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof.
The invention also provides a compound of formula I or formula II or a pharmaceutically acceptable salt thereof for use in therapy, in particular for use in the treatment of migraine. In addition, the present invention provides a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, for use in the prevention of migraine. In addition, the present invention provides the use of a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of migraine.
The invention also provides a pharmaceutical composition comprising a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients. The invention also provides a process for preparing a pharmaceutical composition comprising mixing a compound of formula I or formula II, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients. The invention also encompasses novel intermediates and methods for the synthesis of compounds of formula I and formula II.
The term "C1-C3 alkyl" as used herein refers to methyl, ethyl, propyl and isopropyl.
The term "C3-C5 cycloalkyl" as used herein refers to cyclopropyl, cyclobutyl and cyclopentyl.
The term "treating" as used herein includes inhibiting, slowing, terminating or reversing the progression or severity of an existing symptom or disorder.
The term "prevention" as used herein refers to the protection of a patient susceptible to certain diseases or disorders, such as migraine, but not presently afflicted with the symptoms of the disease or disorder, such as migraine symptoms.
The term "patient" as used herein refers to a mammal, particularly a human.
The term "effective amount" as used herein refers to an amount or dose of a compound of the present invention or a pharmaceutically acceptable salt thereof that provides a desired effect in a patient under diagnosis or treatment after single or multiple dose administration to the patient.
As one skilled in the art, an effective amount can be readily determined by the attending diagnostician using known techniques and by observing results obtained under analogous circumstances. In determining an effective amount for a patient, the attending diagnostician takes into account a number of factors, including but not limited to: the species of the patient; its size, age and general health; the particular disease or disorder involved; the extent or involvement or severity of a disease or disorder; the response of the individual patient; the specific compound administered; the mode of administration; the bioavailability characteristics of the administered formulation; the selected dosing regimen; concomitant drug use; and other related circumstances.
The compounds of the present invention are generally effective over a wide dosage range. For example, daily dosages will generally be in the range of about 0.01 to about 20mg/kg body weight. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed with acceptable side effects, and therefore the aforesaid dosage range is not intended to limit the scope of the invention in any way.
The compounds of the present invention are preferably formulated into pharmaceutical compositions for administration by any route that makes the compounds bioavailable, including oral and transdermal routes. Most preferably, such compositions are for oral administration. Such pharmaceutical compositions and methods for their preparation are well known in the art. (see, e.g., Remington: The Science and Practice of pharmacy (D.B. Troy, Editor, 21 st edition, Lippincott, Williams & Wilkins, 2006).
Compounds of formula I and formula II, or pharmaceutically acceptable salts thereof, are particularly useful in the prophylactic and therapeutic methods of the invention, although certain groups, substituents, and configurations are preferred. The following paragraphs describe such preferred groups, substituents and configurations. Although the present invention contemplates all individual enantiomers and diastereomers as well as enantiomeric mixtures of said compounds including racemates, compounds having the absolute configuration given below are particularly preferred. It will be appreciated that these preferences apply both to the prophylactic and therapeutic methods of the invention and to the novel compounds of the invention.
Preferred are compounds of formula III:
Figure GDA0002319866120000041
or a pharmaceutically acceptable salt thereof.
Also preferred are compounds of formula IV:
Figure GDA0002319866120000042
or a pharmaceutically acceptable salt thereof. Additionally, compounds or salts of formula I, II, III and IV wherein Q is CH are also preferred. Also preferred are compounds or salts of formula I, II, III and IV wherein Y is CH and Z is N. Preference is given to compounds or salts of the formulae I, II, III and IV in which R is C1-C3 alkyl, particularly preferably in which R is methyl.
More preferred are the following compounds:
Figure GDA0002319866120000043
Figure GDA0002319866120000051
and pharmaceutically acceptable salts thereof.
In addition, the following compounds are particularly preferred:
Figure GDA0002319866120000052
and pharmaceutically acceptable salts thereof, with HCl salts and the free base being most particularly preferred.
Furthermore, the following HCl salts are even more preferred:
Figure GDA0002319866120000053
in addition, particularly preferred are the hydrated forms of the above HCl salts, including crystalline forms of the above hydrated forms of the HCl salt, characterized by having a main peak at a diffraction angle 2 θ of 20.7 ° and having one or more peaks selected from 19.8 °, 12.9 °, and 14.0 ° in an X-ray diffraction spectrum; the error in diffraction angle was 0.2 degrees.
In addition, certain intermediates described in the preparations below may contain one or more nitrogen protecting groups. It is understood that the protecting groups may vary as understood by those skilled in the art, depending on the particular reaction conditions and the particular conversion to be carried out. Protection and deprotection conditions are well known to those skilled in the art and are described in the literature (see, e.g., "Greene's Protective Groups in Organic Synthesis", 4 th edition, Peter G.M.Wuts and Theodora W. Greene, John Wiley and Sons, Inc.2007).
The individual isomers, Enantiomers and diastereomers can be separated or resolved by methods such as selective crystallization techniques or chiral chromatography at any suitable point in the synthesis of the Compounds of the invention by one of ordinary skill in the art (see, e.g., j. jacques et al, "eneriomers, racemes, and solutions", John Wiley and Sons, inc.,1981, and e.l. eliel and s.h. Wilen, "Stereochemistry of Organic Compounds", Wiley-Interscience, 1994).
Pharmaceutically acceptable salts of the compounds of the invention, for example, the hydrochloride salt, may be formed, for example, by reacting a suitable free base of a compound of the invention, a suitable pharmaceutically acceptable acid, for example, hydrochloric acid, in a solvent, for example, diethyl ether, under standard conditions well known in the art. In addition, the formation of such salts may occur simultaneously with the deprotection of the nitrogen protecting group. The formation of such salts is well known and understood in the art. See, e.g., Gould, P.L., "Salt selection for basic rules," International Journal of pharmaceuticals, 33: 201-217 (1986); bastin, R.J., et al, "Salt Selection and Optimization Procedures for Pharmaceutical New chemical Entitures," Organic Process Research and Development, 4: 427-435 (2000); and Berge, s.m., et al, "Pharmaceutical Salts," Journal of Pharmaceutical Sciences, 66: 1-19,(1977).
Some abbreviations are defined as follows: "ACN" refers to acetonitrile; "AcOH" refers to glacial acetic acid; "BOP" means (benzotriazol-1-yloxy) tris (dimethylamino) phosphonium hexafluorophosphate; "c-Bu" means cyclobutyl; "c-Pr" means cyclopropyl; "DCM" means dichloromethane; "DIPEA" refers to N, N-diisopropylethylamine; "DMEA" refers to N, N-dimethylethylamine; "DMF" refers to N, N-dimethylformamide; "DMSO" refers to dimethylsulfoxide; "EDCI" refers to 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide; "EDTA" means ethylenediaminetetraacetic acid; "EtOAc" refers to ethyl acetate; "Et" means ethyl; "EtOH" refers to ethanol; "HATU" refers to 1- [ bis (dimethylamino) methylene ] hexafluorophosphate]-1H-1,2, 3-triazolo [4,5-b]Pyridinium 3-oxide; "HPLC" refers to high performance liquid chromatography; "HOAT" refers to 1-hydroxy-7-azabenzotriazole; "HOBt" refers to hydroxybenzotriazole; "hr" means hours; "HTRF" refers to Homogeneous Time resolved fluorescence (Homogeneous Time resolved fluorescence); ' IC50"refers to a concentration of 50% of the maximal inhibitory response of an active agent possible to produce the active agent; "i-Pr" means isopropyl; IPAm "refers to isopropylamine; "kPa" means kilopascals; ' KOtBu "means potassium tert-butoxide; "LAH" refers to lithium aluminum hydride; "LC-ES/MS" refers to liquid chromatography(ii) spray mass spectrometry; "LDA" refers to lithium diisopropylamide; "LDI" refers to laser Doppler imaging; "min" means minutes; "Me" means methyl; "MeOH" refers to methanol; "MTBE" means methyl tert-butyl ether; "NHP" refers to a non-human primate; "NMP" means N-methylpyrrolidone or 1-methylpyrrolidone; "psi" means pounds per square inch; "rpm" means revolutions per minute; "RT" means room temperature; "SEM" refers to the standard error of the mean; "SFC" refers to supercritical fluid chromatography; "SNAR" refers to a nucleophilic aromatic substitution reaction; "T3P" refers to a solution of 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxatriphospha-cyclohexane-2, 4, 6-trioxide; "t-BuOH" means t-butanol; "TEA" refers to triethylamine; "TFA" refers to trifluoroacetic acid; "THF" refers to tetrahydrofuran; "t" sR"refers to retention time; "U/mL" refers to units per milliliter.
The compounds of the present invention or salts thereof may be prepared by a variety of methods known to those skilled in the art, some of which are illustrated in the schemes, preparations and examples below. One skilled in the art recognizes that the specific synthetic steps of each of the routes described can be combined in different ways or in conjunction with steps from different schemes to prepare the compounds of the invention or salts thereof. The product of each step in the following schemes may be recovered by conventional methods well known in the art, including extraction, evaporation, precipitation, chromatography, filtration, trituration, and crystallization. In the following schemes, all substituents are as previously defined unless otherwise indicated. Reagents and starting materials are readily available to those skilled in the art. The following schemes, preparations, examples and assays further illustrate the invention, but should not be construed as limiting the scope of the invention in any way.
Scheme 1
Figure GDA0002319866120000071
In step A of scheme 1, at low temperature, in the presence of about 1.05 equivalents of an organic non-nucleophilic base such as TEA, as appropriateAbout 1 equivalent of 3-bromopyridin-2-amine is slowly treated with a small excess of acryloyl chloride in an organic solvent such as DCM. The reaction mixture was stirred at low temperature for about 2 hr. The reaction is then quenched with an appropriate amount of water, gradually warmed to RT, and the product isolated and purified using standard techniques well known in the art, such as extraction, followed by evaporation of the solvent. For example, with a suitable organic solvent such as DCM and saturated NaHCO3The reaction mixture was worked up with aqueous solution while stirring. The layers were separated, the aqueous layer was extracted with DCM, the organic layers were combined and dried over anhydrous Na2SO4Drying, filtration and concentration under reduced pressure gave the product of step A, N- (3-bromopyridin-2-yl) prop-2-enamide, which was suitable for use without further purification.
In step B of scheme 1, the N- (3-bromopyridin-2-yl) prop-2-enamide prepared in step A is suitably protected in the presence of a strong base in a suitable polar organic solvent. For example, about 1 equivalent of N- (3-bromopyridin-2-yl) prop-2-enamide is added dropwise over 45min at 0 ℃ to a suspension of about 1.5 equivalents of a 60% dispersion of NaH in mineral oil in NMP. Then slowly add about 1.5 equivalents of 2- [ (trimethylsilyl) ethoxy group at 0-5 deg.C]A solution of methyl chloride in NMP was stirred for about 4hr and warmed to about 10 deg.C. Then with saturated NH4The reaction is stopped by aqueous Cl, and the product is isolated and purified using standard techniques well known in the art, such as extraction followed by chromatography. For example, the reaction mixture is treated with a suitable organic solvent such as MTBE and the layers separated; extracting the aqueous layer with MTBE, combining the organic layers, and adding anhydrous Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a crude product in the step B. The crude product is then purified by flash chromatography on silica gel, eluting with a suitable mixture of organic solvents, e.g. hexane/ethyl acetate, to give N- (3-bromopyridin-2-yl) -N- { [2- (trimethylsilyl) ethoxy ] ethanol of step B]Methyl prop-2-enamide.
In step C of scheme 1, about 1 equivalent of N- (3-bromopyridin-2-yl) -N- { [2- (trimethylsilyl) ethoxy ] methyl } prop-2-enamide of step B is treated with about 0.1 equivalent of 2,2' -diazen-1, 2-diylbis (2-methylpropanenitrile and about 1.1 equivalents of tri-N-butyltin hydride under a nitrogen atmosphere in a suitable non-polar solvent such as toluene, then the reaction is heated in a sealed vessel at about 85 deg.C for about 16hr, then the reaction is gradually cooled to RT, the product is isolated and purified using standard techniques well known in the art, such as solvent removal methods followed by chromatography, e.g., evaporation of the reaction mixture under reduced pressure to give the crude product of step C, then the crude product is purified by flash chromatography on silica gel, elution with a suitable mixture of organic solvents, e.g., hexane/ethyl acetate, affords the purified 3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-b ] pyridin-2-one of step C.
Scheme 2
Figure GDA0002319866120000081
In step A of scheme 2, 1- (6-bromopyridin-3-yl) ethanone can be reduced in a stereoselective manner by hydrogenation in the presence of a series of transition metal catalysts. For example, about 0.00075 equivalents of chloro { (R) - (+) -2,2' -bis [ di (3, 5-xylyl) phosphino ] phosphine is used in a suitably sealed and evacuated hydrogenation vessel]-1,1' -binaphthyl } [ (2R) - (-) -1- (4-methoxyphenyl-kC) -3-methyl-1, 2-butanediamine]Ruthenium (II) [ (R) -RUCYTM-XylBINAP]And about 0.0075 equivalents of KOtBu treated about 1 equivalent of 1- (6-bromopyridin-3-yl) ethanone in a suitable polar solvent such as EtOH: 2-propanol (about 1.2mL:1 mL). The system was then charged with hydrogen and stirred at about RT for about 6 hr. The crude product is isolated and purified using standard techniques well known in the art, such as filtration, solvent removal and chromatography. For example, the reaction mixture is filtered and evaporated under reduced pressure to give the crude product of step C. The crude product is then purified by flash chromatography on silica, eluting with a suitable mixture of organic solvents such as DCM/MTBE, to give (1S) -1- (6-bromopyridin-3-yl) ethanol of step A.
In step B of scheme 2, the product of step A, (1S) -1- (6-bromopyridin-3-yl) ethanol is dissolved in a suitable organic solvent, such as DCM, and treated with about 1.3 equivalents of a suitable organic non-nucleophilic base, such as TEA, at about 0 ℃. About 1.2 equivalents of a suitable sulfonylating agent such as methanesulfonyl chloride is added,the product is isolated and purified using standard techniques well known in the art, such as extraction. For example, the reaction mixture is treated with water and the layers are separated; the aqueous layer was extracted 2 times with DCM, the organic layers were combined and saturated NaHCO was used3Washing with aqueous solution, and adding anhydrous Na2SO4Drying, filtration and concentration under reduced pressure gave 1- (6-bromopyridin-3-yl) ethyl (1S) -methanesulfonate from step B, which was used in the next step without further purification.
Scheme 3
Figure GDA0002319866120000091
In step A of scheme 3, about 1.1 equivalents of the product of step C of scheme 1, 3-methyl-1- { [2- (trimethylsilyl) ethoxy ] ethanol]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one and about 1 equivalent of the product of step B, scheme 2, 1- (6-bromopyridin-3-yl) ethyl methanesulfonate, were dissolved in a suitable polar organic solvent such as DMF. Using about 1.2 equivalents of a suitable inorganic base such as Cs at 0 deg.C2CO3And (4) processing the reaction. The reaction mixture was gradually warmed to RT and stirred for about 16 hr. The product is isolated and purified using standard techniques well known in the art, such as extraction followed by flash chromatography. For example, with a suitable organic solvent such as EtOAc and saturated NaHCO3The reaction mixture was worked up with aqueous solution while stirring. The layers were separated, the aqueous layer was extracted with EtOAc, the organic layers were combined and washed with anhydrous Na2SO4Drying, filtering, and concentrating under reduced pressure. Purification of the crude mixture of diastereomers by flash chromatography on silica, eluting with a suitable mixture of organic solvents, e.g., hexane/EtOAc, followed by evaporation of the solvent, affords the purified product of step A, which is (3R) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl]-3-methyl-1- { [2- (trimethylsilyl) ethoxy]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one, the major diastereomer.
In step B of scheme 3, about 1 equivalent of the product of step A, 3R) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl ] -3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-B ] pyridin-2-one, is combined in a suitable anhydrous organic solvent mixture, such as ACN: MeOH (3:2), containing about 0.1 equivalent of a suitable transition metal catalyst, such as palladium (II) acetate, about 0.12 equivalents of a suitable ligand, such as 1,1' -bis (diphenylphosphino) ferrocene, and about 2.6 equivalents of a suitable non-nucleophilic organic base, such as TEA, in a suitable pressure vessel. Sealing, purging with carbon monoxide, pressurizing, and heating to about 100 deg.C for about 3 hr. The reaction mixture is then cooled to RT and the product purified and isolated using standard techniques well known in the art, such as filtration, solvent evaporation and flash chromatography. For example, the reaction is filtered, the filtrate evaporated, and the crude product purified by flash chromatography on silica gel, eluting with a suitable eluent, such as a hexane/EtOAc gradient, to give the purified product of step B, methyl 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-B ] pyridin-3-yl ] ethyl } pyridine-2-carboxylate.
In step C of scheme 3, about 1 equivalent of the product of step B, 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy-1]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid methyl ester is dissolved in a suitable organic solvent, such as DCM, and treated with about 20 equivalents of a strong organic acid, such as TFA. The reaction was stirred at RT for about 19hr and evaporated under reduced pressure. With a suitable organic solvent such as DCM and saturated NaHCO3The aqueous solution treated the residue while mixing. The layers were separated and the aqueous layer was extracted 2 times with DCM. With Na2SO4The combined organic extracts were dried, filtered and concentrated under reduced pressure. The residue is dissolved in a suitable organic solvent, such as MeOH, and a suitable base, such as 1.5 equivalents of ethylenediamine, is added. The reaction was stirred at RT for about 30min, evaporated under reduced pressure, and the product isolated and purified using standard techniques well known in the art, such as extraction methods and chromatography. For example, the reaction residue is evaporated under reduced pressure and diluted with MeOH and a suitable base such as ethylenediamine. The resulting mixture was stirred at RT for about 30min and evaporated under reduced pressure. With a suitable organic solvent such as DCM and saturated NaHCO3The aqueous solution treated the residue while mixing. The layers were separated and the aqueous layer was extracted 2 times with DCM. With Na2SO4Drying the combined organic extracts, filtering, and concentrating under reduced pressure to obtain step CThe crude product of (1). The crude product is then purified by flash chromatography on silica gel, eluting with a suitable organic eluent, e.g., a DCM/EtOAc gradient, to provide the purified product of step C, 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ]]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid methyl ester.
In step D of scheme 3, about 3 equivalents of a suitable inorganic base such as LiOH are added to about 1 equivalent of the product of step C, 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] in a suitable solvent mixture such as THF/water (5:1)]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid methyl ester. The reaction mixture was stirred at RT for about 2hr and partitioned between a suitable organic solvent and aqueous acid such as DCM and 1M HCl. Then saturated NaHCO3Basifying the mixture to pH-2 with an aqueous solution and mixing with a suitable organic solvent such as CHCl32-propanol (3:1) was extracted about 5 times. With Na2SO4The combined organic extracts were dried, filtered, and concentrated under reduced pressure to give the product of step D, 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b [ ] -]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid, which was used in the next step without purification.
In scheme 3, step E, the product of step D, 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] -can be made using standard amidation synthesis methods well known in the art]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid was coupled with different amines or amine hydrochloride salts. For example, about 1 equivalent of the product of step E can be combined with about 1.7 equivalents of 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride, 1.7 equivalents of 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxane-2, 4, 6-trioxane and 5 equivalents of TEA in a suitable solvent such as DMF. The reaction mixture was stirred at RT for about 17hr, after which the product was isolated and purified using techniques well known in the art, such as extraction and chromatography. For example, saturated NaHCO3An aqueous solution is added to the reaction mixture, which is then extracted with a suitable organic solvent, such as DCM. With Na2SO4The combined organic extracts were dried, filtered, and concentrated under reduced pressure to give the crude product of step E. The crude product can then be purified by flash chromatography on silica gel using a suitable eluent, for exampleElution with a DCM/MeOH gradient afforded the purified product of step E, N- [ (2, 6-dimethylpyridin-4-yl) methyl]-5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxamide.
Scheme 4
Figure GDA0002319866120000121
In step A of scheme 4, about 1.0-1.2 equivalents of Zn (CN)2To a solution of 4-bromo-2, 6-lutidine in a suitable polar organic solvent such as DMF containing about 5-10 mol% of a suitable transition metal catalyst/ligand complex such as tetrakis (triphenylphosphine) palladium (0). After heating for about 5-18hr, the reaction mixture is cooled to RT and the product isolated and purified using standard techniques well known in the art, such as extraction, followed by solvent evaporation or chromatography. For example, with a suitable organic solvent such as EtOAc and NH4The reaction mixture was treated with aqueous OH while mixing. The layers were separated, the aqueous layer was extracted with EtOAc, the organic layers were combined and washed with anhydrous Na2SO4Drying, filtering and concentrating under reduced pressure to obtain the crude product of the step A. The crude product can then be purified by flash chromatography on silica, eluting with a suitable mixture of organic solvents such as hexane/ethyl acetate, to give the product 4-cyano-2, 6-lutidine of step a. Alternatively, with a suitable organic solvent such as MTBE followed by an aqueous alkaline (pH 10) solution such as 30% NH4The crude reaction mixture was diluted with OH, the layers were separated, the aqueous phase was extracted with MTBE and 10% NH4The combined organic extracts were washed with OH and anhydrous Na2SO4Drying, filtration and concentration under reduced pressure gave the product 4-cyano-2, 6-dimethylpyridine of step a, which was suitable for use without further purification.
In scheme 4, step B, the product of step A, 4-cyano-2, 6-lutidine, can be reduced by a variety of methods well known in the art, for example, using a reducing agent such as LiBH4Or NaBH4Reduction by chemical hydrogenation or using transition metals, e.g. Pd (OH) on carbon2Or Pd hydrogenation. Alternatively, the hydrogenation may be inorganicIn the presence of aqueous acid or in a suitable organic solvent such as THF or DMF to give the reduced product as the HCl salt. For example, the product of step A, 4-cyano-2, 6-lutidine, is dissolved in excess HCl in water or 1, 4-di-HCl
Figure GDA0002319866120000132
The alkane solution is dissolved in a suitable organic solvent such as MeOH or EtOH in the presence of 5-10% Pd/C. The reaction mixture was hydrogenated at RT overnight at a pressure of about 60 psi. The mixture was filtered and the filtrate was concentrated to give the crude product of step B. The product of step B can then be precipitated by methods known to those skilled in the art, for example by grinding, crystallization or recrystallization. For example, the crude product of step B can be treated with a boiling EtOH/EtOAc mixture until dissolved; the product was then cooled by crystallization and collected by filtration to give the product 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride from step B. Alternatively, the crude product may be suspended in a mixture of MeOH/MTBE and the resulting solid, product 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride of step B, collected by filtration.
Scheme 5
Figure GDA0002319866120000131
In step A of scheme 5, about 1 equivalent of 3-methyl-1, 3-dihydropyrrolo [2,3-b ] is added]Pyridin-2-one and about 1.05 equivalents of the product of scheme 2, step B, 1- (6-bromopyridin-3-yl) ethyl methanesulfonate were dissolved in a suitable polar organic solvent such as DMF. Using about 1-1.2 equivalents of a suitable inorganic base such as Cs at 0 deg.C2CO3And (4) processing the reaction. The reaction mixture was gradually warmed to RT and stirred for about 16-48 hr. The product is isolated and purified using standard techniques well known in the art, such as extraction methods. For example, the reaction mixture is treated with a suitable organic solvent such as MTBE and water while mixing. Separating the layers, extracting the aqueous layer with MTBE, combining the organic layers, and adding anhydrous Na2SO4Drying, filtering and concentrating under reduced pressure to obtain the crude product (3R) -3- [ (1R) -1- (6-bromo-3-pyridyl) ethyl of step A]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one and (3S) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one as a mixture of diastereomers.
In step B of scheme 5, one skilled in the art will recognize that the diastereomeric product of step A can be resolved by chiral chromatography or by chiral salts such as with enantiomerically pure mandelic, tartaric, or camphorsulfonic acids among other possibilities well known in the art. More specifically, a diastereomeric mixture of the product from step A, which is (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one and (3S) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one, may be dissolved in a suitable organic solvent, such as EtOAc, and treated with 1-1.5 equivalents of (1S) - (+) -10-camphorsulfonic acid. The reaction mixture may be heated for about 30min, cooled to RT with stirring overnight, and the resulting product collected by methods well known in the art, e.g., by vacuum filtration, to give the product of step B, (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-B ] pyridin-2-one, (1S) - (+) -10-camphorsulfonate, with high diastereomeric purity.
In step C of scheme 5, the product (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl) from step B can be reacted under conditions well known in the art]-3-methyl-1H-pyrrolo [2,3-b]The pyridine-2-one, (1S) - (+) -10-camphorsulfonate is isolated as the free base, for example by extraction from an aqueous alkaline solution, which can be crystallized from a suitable organic solvent. Specifically, (3R) -3- [ (1R) -1- (6-bromo-3-pyridyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one, (1S) - (+) -10-Camphorsulfonate dissolved in NaHCO3Aqueous solution, extracted several times with a suitable organic solvent such as EtOAc. The combined organic extracts were washed with saturated aqueous NaCl and Na2SO4Drying, filtration and evaporation of the filtrate under reduced pressure gave the free base as a residue which was crystallized from MeOH and collected by filtration to give the product (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl of step C]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one.
In Steps D and E of scheme 5, strips are well described in the artThe product of step C, namely (3R) -3- [ (1R) -1- (6-bromo-3-pyridyl) ethyl, is catalyzed by a transition metal in a suitable organic solvent]-3-methyl-1H-pyrrolo [2,3-b]Carbonylation of the pyridin-2-one, optionally isolation of the carbonylation product (step D), followed by direct amidation with an amine under amidation conditions well known in the art, yields the amide product of step E. More specifically, the product of step C, (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl, is reacted in a sealed pressurized reaction vessel under an atmosphere of CO at 60psi]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one dissolved in a solution containing about 1.1 equivalents of phenol, about 0.01 equivalents of Pd (OAc)2About 0.01 equivalents of 4, 5-bis (diphenylphosphino) -9, 9-dimethylxanthene and about 5 equivalents of TEA in toluene. The reaction mixture was heated at about 85 ℃ overnight, cooled slightly, and about 1.1 equivalents of 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride was added to the product containing the unseparated step D, 5- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] to]Pyridin-3-yl]Ethyl radical]Pyridine-2-carboxylic acid phenyl ester. The reaction mixture was resealed and heated to about 120 deg.C for about 1 hr. The product of step E may be isolated and purified using standard techniques well known in the art, such as filtration followed by chromatography. Specifically, the reaction mixture is cooled, diluted with a suitable organic solvent such as EtOAc, filtered through a bed of celite, and the filtrate is concentrated under reduced pressure to provide the crude product of step E. The crude product is then purified by flash chromatography on silica gel, eluting with a suitable eluent, such as a mixture of acetone in hexane, to give the purified product of step E, N- [ (2, 6-dimethylpyridin-4-yl) methyl]-5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxamide.
Scheme 6
Figure GDA0002319866120000151
In Steps A-E of scheme 6, various substituted {4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzoyl } amides can be prepared by methods analogous to those described in scheme 3. The requisite [ (1S) -methanesulfonic acid 1- (4-bromophenyl) ethyl ] ester of scheme 6, step A, was prepared from (1S) -1- (4-bromophenyl) ethanol (Accel Pharmtech) under conditions analogous to those described in scheme 2, step B. This mesylate is treated with the product 3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-B ] pyridin-2-one of scheme 1 step C under conditions analogous to those described in scheme 3 step A to give the product (3R) -3- [ (1R) -1- (4-bromophenyl) ethyl ] -3-methyl-1- (2-trimethylsilylethoxymethyl) pyrrolo [2,3-B ] pyridin-2-one of scheme 6 step B. In scheme 6, step C, (3R) -3- [ (1R) -1- (4-bromophenyl) ethyl ] -3-methyl-1- (2-trimethylsilylethoxymethyl) pyrrolo [2,3-B ] pyridin-2-one can be carbonylated under conditions analogous to those described in scheme 3, step B to give methyl 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- (2-trimethylsilylethoxymethyl) pyrrolo [2,3-B ] pyridin-3-yl ] ethyl } benzoate, which can then be deprotected to the lactam nitrogen by analogous methods to those described in scheme 3, step C to give the product of scheme 6, step D, 4- [ (1R) -1- [ (3R) -3R ] -3 -methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzoic acid. Subsequent saponification in step E of scheme 6 under conditions analogous to those described in scheme 3, step D can afford 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid.
Scheme 7
Figure GDA0002319866120000161
In scheme 7, step A, 4-chloro-7- (2, 4-dimethoxybenzyl) -5, 7-dihydro-6H-pyrrolo [2,3-d ] can be performed under basic conditions using a variety of conditions well described in the art]Pyrimidin-6-one (US 2010/0120801) is alkylated. For example, about 1 equivalent of 4-chloro-7- (2, 4-dimethoxybenzyl) -5, 7-dihydro-6H-pyrrolo [2,3-d ] can be added]Pyrimidin-6-one is dissolved in a suitable polar organic solvent such as DMF, slowly treated with a suitable strong inorganic base such as 1 equivalent NaH, followed by treatment with a suitable alkyl halide such as 1 equivalent CH3And I, processing. The reaction mixture was stirred for about 30 min. Using standard techniques well known in the artThe product is isolated and purified by techniques such as extraction and chromatography. For example, with saturated NH4The reaction mixture was diluted with aqueous Cl and extracted with EtOAc. Separating the layers, separating the organic layer, and adding Na2SO4Drying, filtering, and concentrating under reduced pressure. The resulting residue is purified by flash chromatography on silica, eluting with a suitable mixture of organic solvents such as hexane/EtOAc, and then evaporating the solvent to provide the product 4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] of scheme 7, step A]The product of step A, scheme 7, 4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] can be reacted with the desired α -methylpyridyl-or α -methylphenyl-methanesulfonate under conditions analogous to those described in step A, scheme 3, or step B, scheme 6, respectively]Pyrimidin-6-one alkylation to give the product of scheme 7, step B (X ═ N, CH). The product of step B, scheme 7, (X ═ N, CH) can be dechlorinated under hydrogenation conditions well understood in the art. For example, the product of step B of scheme 7 (X ═ N, CH) is dissolved in a suitable polar organic solvent such as MeOH, treated with a suitable transition metal catalyst suitable for hydrogenation, such as 5% Pd on carbon, and hydrogenated under conditions well known in the art, such as under a hydrogen atmosphere at pressures of 20 to 60 psi. The reaction mixture was filtered and the filtrate was evaporated under reduced pressure. The crude dechlorinated product may be used as such (X ═ N) or may be purified using standard techniques well known in the art, such as extraction and chromatography (X ═ CH). For example, with saturated NaHCO3The crude filtrate is diluted with aqueous solution, extracted with a suitable organic solvent such as DCM and the layers separated. With Na2SO4The organic extract is dried, concentrated under reduced pressure, and the resulting residue is purified by flash chromatography on silica, eluting with a suitable organic solvent mixture such as hexane/EtOAc, to afford, after evaporation of the solvent, the dechlorinated product of scheme 7, step C (X ═ CH). The lactam nitrogen of the product of step C of scheme 7 can be exposed by conditions well understood in the art. For example, about 1 equivalent of the product of step C of scheme 7 is dissolved in a mixture of a suitable high boiling organic solvent such as anisole and a strong organic acid such as TFA. Heating the reaction mixture under heat or microwave for about 2-18hr, and extracting to obtain crude productE.g. with saturated NaHCO3The reaction mixture is diluted with a mixture of aqueous solution and a suitable organic solvent, such as DCM. Separating the layers with Na2SO4The organic layer was dried, filtered and the crude product was purified by flash chromatography on silica gel eluting with a suitable mixture of organic solvents such as DCM/EtOAc/MeOH (X ═ N) or DCM/EtOAc (X ═ CH) to afford the product of scheme 7, step D; saponification under conditions analogous to those described in scheme 3, step D (X ═ N) or scheme 6, step E (X ═ CH) affords the desired acidic product of scheme 7, step E.
Scheme 8
Figure GDA0002319866120000181
Scheme 8 describes the preparation of 2-substituted-6-methyl-pyridylmethylamines. In scheme 8, step A, the 2-chloropyridine is converted to a 2-alkylpyridine using a Grignard reagent, an alkyllithium, an alkylborate or an alkylzinc reagent, as will be appreciated by those skilled in the art. For example, in a suitable polar solvent such as NMP or 1, 4-bis
Figure GDA0002319866120000182
In an alkane or in a biphasic mixture of a suitable organic solvent containing water, e.g., toluene, benzene or DMF, in about 0.1-0.2 equivalents of a transition metal catalyst such as iron (III) acetoacetate (R ═ Et), Pd (OAc)2In the presence of a suitable phosphine ligand, for example tricyclohexylphosphine tetrafluoroborate or [1,1' -bis (diphenylphosphino) -ferrocene]Treating about 3.0-3.6 equivalents of 2-chloro-6-methylisonicotonitrile (Bioorganic) with about 1.0-1.5 equivalents of an appropriately substituted grignard reagent, alkyl borate or alkyl zinc reagent in the presence of palladium (II) dichloride (R ═ i-Pr, c-Bu, cyclopentyl) at about room temperature to about 120 ℃&Medicinal Chemistry Letters,20(2), 576-580; 2010) to give the crude 2-alkyl product of scheme 8, step A, which can be isolated and purified under conditions well known in the art, such as extraction and chromatography. For example, the reaction is diluted with water, filtered through a bed of celite and the filtrate is extracted with a suitable organic solvent such as EtOAc or DCM. With Na2SO4The organic extracts were dried, filtered, concentrated under reduced pressure, and purified by flash chromatography on silica gel, eluting with hexane or heptane/EtOAc, to give the desired product, 2-alkyl-6-methyl-4-pyridinecarbonitrile, step a of scheme 8. The partial reduction of the carbonitrile to methylamine may be carried out under a range of conditions well understood in the art. For example, an excess of Raney nickel may be used under a hydrogen atmosphere of 20 to 60psi in a suitable polar solvent mixture such as NH3Is treated with the desired approximately 1 equivalent of the product 2-alkyl-6-methyl-4-pyridinecarbonitrile of scheme 8, step A. The reaction mixture may be filtered, concentrated, and the resulting residue triturated sequentially with an appropriate mixture of organic solvents such as toluene, ACN, MeOH/toluene, and ACN/toluene, followed by filtration to give the appropriately substituted (2-alkyl-6-methyl-4-pyridinyl) methylamine as the dihydrochloride salt. Alternatively, the resulting crude product may be isolated and purified under conditions well known in the art, such as extraction and chromatography, to give the appropriately substituted (2-alkyl-6-methyl-4-pyridyl) methylamine as the free base.
Scheme 9
Figure GDA0002319866120000191
Scheme 9 describes the preparation of compounds of formula IV, wherein the appropriate carboxylic acid can be coupled with the appropriate amine under a range of amide coupling conditions well known in the art. For example, the amide coupling reaction can be carried out in a manner similar to that described in scheme 3, step E, or using coupling reagents such as EDCI, HOBt, HOAT, HATU, or T3P, which are well described in the literature.
Scheme 10
Figure GDA0002319866120000192
Scheme 10 describes the preparation of 6- (aminomethyl) -4-methyl-pyridine-2-carbonitrile dihydrochloride. In scheme 10, step a, the reduction of 2-pyridine-carboxylic acid ethyl ester can be achieved under a range of methods well described in the art. For example, with about 1.7 equivalents of sodium borohydride in EtOH at RTLeaving about 1 eq of ethyl 6-chloro-4-methylpyridine-2-carboxylate (Y ═ CH, Z ═ N) to give the product of step a of scheme 10 (Y ═ CH, Z ═ N) (6-chloro-4-methyl-2-pyridinyl) methanol, which is suitable for use without further purification. The person skilled in the art knows to carry out the halogenation under various halogenation conditions to give alkyl halides. For example, at about RT to reflux in a suitable organic solvent such as DCM or CHCl3The desired product of scheme 10, step B, (Y ═ CH, Z ═ N) 2-chloro-6- (chloromethyl) -4-methyl-pyridine can be obtained by treatment of about 1 equivalent of the product of scheme 10, step a, (6-chloro-4-methyl-2-pyridinyl) methanol with about 2 equivalents of thionyl chloride and evaporation of the solvent, which is suitable for use without further purification. The product of scheme 10, step B, (Y ═ CH, Z ═ N) 2-chloro-6- (chloromethyl) -4-methyl-pyridine can be treated with a variety of protected amines suitable to withstand additional functionalization. For example, about 1 equivalent of potassium phthalimide may be treated with the product of scheme 10, step B, (Y ═ CH, Z ═ N) 2-chloro-6- (chloromethyl) -4-methyl-pyridine in a suitable polar solvent such as DMF. Subsequent dilution with water affords the solid product of scheme 10, step C, (Y ═ CH, Z ═ N)2- [ (6-chloro-4-methyl-2-pyridinyl) methyl]Isoindoline-1, 3-dione, which can be isolated by methods well known in the art, such as filtration. The product of scheme 10 step C (Y ═ CH, Z ═ N)2- [ (6-chloro-4-methyl-2-pyridyl) methyl]The chloride moiety of isoindoline-1, 3-diones can be displaced by various nucleophiles well described in the literature, for example, by SNAR reactions or by transition metal mediated methods. For example, at about 0.05 equivalent of [1,1' -bis (diphenylphosphino) ferrocene ] dichloride]Treating about 1 equivalent of the product of step C, scheme 10, (Y ═ CH, Z ═ N)2- [ (6-chloro-4-methyl-2-pyridinyl) methyl ] 2- [ (6-chloro-N-methyl-2-pyridinyl) methyl ] with about 0.75 equivalents of zinc cyanide in the presence of palladium (II) and about 0.25 equivalents of elemental zinc in a suitable polar organic solvent such as DMF or DMSO under heating at 100 ℃ 140 ℃]Isoindoline-1, 3-dione. The product of such a conversion can be isolated and purified by standard techniques well known in the art, such as extraction and chromatography, as is known to those skilled in the art. For example, the cooled reaction mixture can be diluted with water, extracted with a suitable solvent such as DCM or EtOAc, followed by NH4OH and saturated aqueous NaCl solutionWashing with Na2SO4Or MgSO 24The organic extract is dried. The crude product obtained can be flash chromatographed on silica gel, eluting with a suitable mixture of organic solvents, e.g., hexane/ethyl acetate, to provide the product of scheme 10, step D (Y ═ CH, Z ═ N)6- [ (1, 3-dioxoisoindolin-2-yl) methyl)]-4-methyl-pyridine-2-carbonitrile. Removal of the amine protecting group can be accomplished by one skilled in the art. For example, about 1 equivalent of the product of step D of scheme 10 (Y ═ CH, Z ═ N)6- [ (1, 3-dioxoisoindolin-2-yl) methyl) is treated with about 2 equivalents of hydrazine hydrate in a suitable polar organic solvent such as EtOH at reflux]-4-methyl-pyridine-2-carbonitrile, which gives the crude deprotected amine after evaporation of the solvent. The crude amine can then be isolated and purified by standard methods well known in the art, such as selective cation exchange and salt preparation. For example, the crude amine can be passed through an SCX column using NH3Elution with MeOH mixture; the methanolic fraction of the amine can be evaporated, the resulting residue redissolved in MeOH, and treated with 2-10 equivalents of HCl in a suitable organic solvent such as Et2O or 1, 4-di
Figure GDA0002319866120000201
The resulting solution was treated in an alkane to yield solid 6- (aminomethyl) -4-methyl-pyridine-2-carbonitrile dihydrochloride salt after collection by filtration. Compounds in which Y ═ N and Z ═ CH can be synthesized by analogous methods.
Preparation examples and examples
The following preparations and examples further illustrate the invention and represent typical syntheses of the compounds of the invention. Reagents and starting materials are readily available or can be readily synthesized by those skilled in the art. It is understood that the preparations and examples are given by way of illustration and are not limiting of the invention, and that various modifications may be effected therein by those skilled in the art.
The R-or S-configuration of the compounds of the invention can be determined by standard techniques such as X-ray analysis and correlation with chiral-HPLC retention time.
Can use
Figure GDA0002319866120000211
The HP1100 liquid chromatography system performed LC-ES/MS. Electrospray mass spectrometry measurements (acquired in positive and/or negative mode) were performed using a mass selective detector quadrupole mass spectrometer interfaced to HP1100 HPLC. LC-MS conditions (low pH): column:
Figure GDA0002319866120000212
Figure GDA0002319866120000213
NX C182.1X 50mm3.5 μm; gradient: 5-100% B, 3min, then 100% B, 0.75min, or 5-95% B, 1.5min, then 95% B, 0.25 min; column temperature: 50 deg.C +/-10 deg.C; flow rate: 1.2 mL/min; solvent A: deionized water containing 0.1% HCOOH; solvent B: CAN containing 0.1% formic acid; the wavelength is 214 nm. Alternative LC-MS conditions (high pH): column:
Figure GDA0002319866120000214
MS C18 column 2.1X 50mm,3.5 μm; gradient: 5% solvent a, 0.25min, gradient 5% -100% solvent B, 3min, and 100% solvent B, 0.5min, or 10% -100% solvent B, 3min, and 100% solvent B, 0.75min, or 5-95% B, 1.5min, then 95% B, 0.25 min; column temperature: 50 deg.C +/-10 deg.C; flow rate: 1.2 mL/min; solvent A: 10mM NH4HCO3pH 9; solvent B: ACN; wavelength: 214 nm.
Using mass-selective mass spectrometers equipped with
Figure GDA0002319866120000215
Of automatic samplers/fraction collectors
Figure GDA0002319866120000216
1200LC-ES/MS performed preparative reverse phase chromatography. High pH method at 75X 30mm with 10X 20mm guard column
Figure GDA0002319866120000217
On a 5 μ particle size column. The flow rate was 85 mL/min. The eluent is 10mM ammonium bicarbonate (pH 10)Acetonitrile solution.
CDCl using Bruker AVIII HD 400MHz NMR spectrophotometer or Varian VNMRS 400MHz NMR spectrophotometer3Or (CD)3)2NMR spectra were obtained as SO solution, reported in ppm, using residual solvent [ CDCl ]3,7.26ppm;(CD3)2SO,2.05ppm]As a reference standard. When reporting peak diversity, the following abbreviations may be used: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br-s (broad singlet), dd (doublet), dt (doublet triplet). When reported, the coupling constants (J) are reported in Hertz (Hz).
X-ray powder diffraction: XRD spectra of crystalline solids
Figure GDA0002319866120000222
) And Vantec detector on a Bruker D4 energy X-ray powder diffractometer, operating at 50kV and 50 mA. The sample was scanned between 4 and 40 ° in 2 θ, with a step size of 0.009 ° in 2 θ, a scan speed of 0.5 sec/step, 0.6mm divergence, 5.28 fixed anti-scatter and 9.5mm detector slit. The dried powder was mounted on a quartz sample holder and a smooth surface was obtained using a glass slide. The crystalline form diffraction spectra were collected at ambient temperature and relative humidity. It is well known in the crystallography art that for any given crystalline form, the relative intensities of the diffraction peaks may vary due to preferred orientations resulting from factors such as crystal morphology and habit. In the presence of the influence of preferred orientation, the peak intensity changes, but the characteristic peak position of the polymorph does not. See, for example, The United States Pharmacopeia #23, National Formulary #18, pages 1843-1844, 1995. Furthermore, it is also well known in the crystallography art that for any given crystalline form, the angular peak positions may vary slightly, e.g., the peak positions may shift due to changes in temperature or humidity at which the sample is analyzed, sample displacement, or the presence or absence of an internal standard. In the present case, peak position variations of ± 0.2 in 2 θ will take these potential variations into account without preventing a clear identification of the given crystalline form. Confirmation of the crystal form may be based on any unique combination of distinct peaks (in ° 2 θ), typically more pronouncedProminent peaks. The crystal form diffraction spectra collected at ambient temperature and relative humidity were adjusted based on NIST 675 standard peaks at 8.85 and 26.77 degrees 2-theta.
Preparation example 1
N- (3-bromopyridin-2-yl) propan-2-enamides
Figure GDA0002319866120000221
Scheme 1, step a: a solution of 3-bromopyridin-2-amine (100.0g,578.0mmol) and TEA (82mL, 588.0mmol) in DCM (2.8L) was cooled to-78 ℃. Acryloyl chloride (47.5mL,584.0mmol) in DCM (200mL) was added dropwise over 2 hr. The reaction mixture was stirred at-75 ℃ for 2 hr. Adding water, then adding saturated NaHCO3Aqueous solution, and the layers were separated. With Na2SO4The organic layer was dried, filtered, and concentrated under reduced pressure to give a solid. To the solid was added DCM (200mL) and MTBE (600mL), and the mixture was concentrated under reduced pressure to about 300 mL. The resulting off-white solid was filtered and dried under vacuum overnight to give the title compound (97.1g, 74% yield). LC-ES/MS (m/z,79Br/81Br):227.0/229.0(M+H).
preparation example 2
N- (3-bromopyridin-2-yl) -N- { [2- (trimethylsilyl) ethoxy ] methyl } prop-2-enamide
Figure GDA0002319866120000231
Scheme 1, step B: n- (3-bromopyridin-2-yl) prop-2-enamide (42g,185.0mmol) in NMP (800mL) was added over 45min to a suspension of NaH (60% in mineral oil, 11.1 g,277.0mmol) in NMP (800mL) at 0 deg.C and stirred for 30min at 0-5 deg.C. Adding 2- [ (trimethylsilyl) ethoxy group dropwise at 0-5 deg.C for 30min]Methyl chloride (50mL,280.0 mmol). The reaction mixture was stirred at 0 ℃ for 4hr and then warmed to 10 ℃. After 15min at 10 ℃, saturated NH is added4Aqueous Cl and water, and the mixture was extracted with MTBE. The combined organic layers were washed successively with water and then with saturated aqueous NaCl and Na2SO4The mixture is dried and then is dried,filtration and concentration under reduced pressure gave a residue. The residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 1:0-3:1) to give the title compound (48.0g, 73% yield) after evaporation of the solvent. LC-ES/MS (m/z,79Br/81Br):357.0/359.0(M+H).
preparation example 3
3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-b ] pyridin-2-one
Figure GDA0002319866120000232
Scheme 1, step C: to a solution of N- (3-bromopyridin-2-yl) -N- { [2- (trimethylsilyl) ethoxy ] methyl } prop-2-enamide (2.17g,6.1mmol) in toluene (61mL) were added 2,2' -diazene-1, 2-diylbis (2-methylpropanenitrile) (0.100g,0.61mmol) and tri-N-butyltin hydride (1.94g, 6.7 mmol). The reaction mixture was purged with nitrogen and heated to 85 ℃ for 16 hr. The mixture was cooled to RT and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 1:0-1:1) to give the title compound (1.08g, 64% yield) after evaporation of the solvent. LC-ES/MS (m/z): 279.0(M + H).
Preparation example 4
(1S) -1- (6-bromopyridin-3-yl) ethanol
Figure GDA0002319866120000241
Scheme 2, step a: reacting under nitrogen, chloro { (R) - (+) -2,2' -bis [ di (3, 5-xylyl) phosphino]-1,1' -binaphthyl } [ (2R) - (-) -1- (4-methoxyphenyl-kC) -3-methyl-1, 2-butanediamine]Ruthenium (II) (103mg,0.087mmol) and KOtA solution of Bu (1.0M solution of t-BuOH, 0.88 mL,0.88mmol) in anhydrous 2-propanol (15mL) was added to a solution of 1- (6-bromopyridin-3-yl) ethanone (23.5g,117.0mmol) in anhydrous EtOH (100 mL)/anhydrous 2-propanol (85mL) in a 600mL Parr autoclave under nitrogen. The autoclave was sealed, evacuated, pressurized to 207kPa with hydrogen and stirred at RT for about 6 hr. Vacuum concentration reactionThe mixture was allowed to stand to give a solid residue which was dried under vacuum overnight. The residue was purified by flash chromatography on silica eluting with DCM/MTBE (gradient 9:1-3:1) to give the title compound (23.7g, 94% yield). LC-ES/MS (m/z,79Br/81br): 202.0/204.0(M + H). Chiral HPLC showed 99.3% ee; t is tR6.32min [254 nm; an LC column:
Figure GDA0002319866120000243
OD-H4.6 x150 mm; 5.0 μ L injection; 10% 2-propanol in heptane (containing 0.2% DMEA); column temperature: 25 ℃; flow rate: 1.0mL/min]。
Preparation example 5
(1S) -methanesulfonic acid 1- (6-bromopyridin-3-yl) ethyl ester
Figure GDA0002319866120000242
Scheme 2, step B: to a stirred solution of (1S) -1- (6-bromopyridin-3-yl) ethanol (3.00g,14.8mmol) and TEA (2.69mL,19.3mmol) in DCM (30mL) was added methanesulfonyl chloride (1.38mL,17.8mmol) at 0 deg.C. After 2hr at 0 deg.C, water and DCM were added and the layers were separated. The aqueous layer was extracted with DCM. The organic layers were combined and successively saturated NaHCO3Aqueous solution and saturated NaCl, washed with Na2SO4Drying; filtration and concentration under reduced pressure gave the title compound (4.13g, 99% yield). LC-ES/MS (m/z,79Br/81Br):280.0/282.0(M+H).1H NMR (CDCl3)δ1.74(d,J=6.6Hz,3H),2.93(s,3H),5.76(q,J=6.6Hz,1H),7.54(d, J=8.3Hz,1H),7.62(dd,J=2.5,8.3Hz,1H),8.41(d,J=2.5Hz,1H).
preparation example 6
(3R) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl ] -3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-b ] pyridin-2-one (major diastereomer) and (3S) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl ] -3-methyl-1- { [2- (trimethylsilyl) ethoxy ] methyl } -1, 3-dihydro-2H-pyrrolo [2,3-b ] pyridin-2-one (minor diastereomer).
Figure GDA0002319866120000251
Scheme 3 step a: to a stirred mixture of 3-methyl-1- { [2- (trimethylsilyl) ethoxy ] at 0 ℃ under nitrogen]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one (4.99g, 90% purity, 16.1mmol) and a solution of 1- (6-bromopyridin-3-yl) ethyl (1S) -methanesulfonate (4.12g,14.7mmol) in DMF (74mL) were added Cs2CO3(5.75g,17.6 mmol). The reaction mixture was purged thoroughly with nitrogen and gradually warmed to RT. After stirring at RT for 16hr, EtOAc and saturated aqueous NaHCO solution are added3The layers were separated. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with Na2SO4Dried, filtered and concentrated under reduced pressure to give a red oil. The crude product was purified by flash chromatography on silica eluting with hexane/EtOAc (9:1-3:2) to give, after evaporation of the solvent, the title compound (4.61g, 65% yield) as the major enantiomer, which eluted first, and the second minor enantiomer (3S) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl) which eluted second]-3-methyl-1- { [2- (trimethylsilyl) ethoxy]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one (0.34g, 5% yield).
The major diastereomer: LC-ES/MS (m/z,79Br/81Br):462.0/464.0(M+H).1H NMR(CDCl3)δ-0.02(s,9H),0.90-1.03(m,2H),1.20(d,J=7.2Hz,3H),1.35 (s,3H),3.30(q,J=7.2Hz,1H),3.60-3.64(m,2H),5.19-5.25(m,2H),6.94 (dd,J=5.2,7.3Hz,1H),7.05(dd,J=1.6,7.3Hz,1H),7.33(dd,J=2.5,8.2 Hz,1H),7.42(d,J=8.2Hz,1H),8.10(d,J=2.5Hz,1H),8.21(dd,J=1.6, 5.2Hz,1H).
minor diastereomer: LC-ES/MS (m/z,79Br/81Br):462.0/464.0(M+H).1H NMR(CDCl3)δ-0.03(s,9H),0.81-0.96(m,2H),1.44(d,J=7.2Hz,3H), 1.47(s,3H),3.30(q,J=7.2Hz,1H),3.34-3.40(m,2H),4.96-5.02(m,2H), 7.03(dd,J=5.3,7.3Hz,1H),7.06(dd,J=2.5,8.3Hz,1H),7.21(d,J=8.3 Hz,1H),7.61(dd,J=1.6,8.2Hz,1H),7.92(d,J=2.5Hz,1H),8.22(dd,J= 1.6,5.3Hz,1H).
preparation example 7
5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000261
Scheme 3, step B: palladium (II) acetate (227mg,0.961mmol), 1' -bis (diphenylphosphino) ferrocene (670mg,1.17mmol), (3R) -3- [ (1R) -1- (6-bromopyridin-3-yl) ethyl]-3-methyl-1- { [2- (trimethylsilyl) ethoxy]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one (4.61g,9.58mmol), anhydrous MeOH (40mL), anhydrous ACN (60mL), and TEA (3.5mL,25mmol) were combined in a 300mL Parr autoclave with mechanical stirrer. The autoclave was sealed, purged with CO, pressurized to 689kPa with CO, heated to 85 ℃ while stirring. After 3hr at 100 ℃, the reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure to give a solid residue. The residue was suspended in EtOAc (200mL) and filtered again. The filtrate was concentrated under reduced pressure to give an orange residue. The orange residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 7:3-8:2) and the desired fractions evaporated to afford the title compound (3.69g, 86% yield). LC-ES/MS (m/z): 442.2(M + H). Passing through chiral SFC (column: Lux Cellulose-4; eluent: 20:80, MeOH: CO)2(ii) a Flow rate: 5mL/min, UV225nm) showed 98.3% ee, corresponding to the first eluting isomer, tR=1.33min.
Preparation example 8
5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000271
Scheme 3, step C: to 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy at RT]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid methyl ester (3.67g,8.21mmol) in DCM (27)4mL) was added TFA (12.4 mL,164.0 mmol). After 19hr, the solution was concentrated under reduced pressure to give a residue. The residue was taken up in DCM and saturated NaHCO3The aqueous solution was partitioned between and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. MeOH (109mL) and ethylenediamine (0.826mL,12.3mmol) were added to the residue. After 30min, the solution was concentrated under reduced pressure to give a residue. DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The residue was purified by flash chromatography on silica eluting with DCM/EtOAc (gradient 1:0-0:1) to give the title compound (1.80g, 70% yield) after evaporation of the solvent. LC-ES/MS (m/z): 312.0(M + H).
Preparation example 9
5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxylic acid
Figure GDA0002319866120000272
Procedure 3, step D: to 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] at RT]Pyridin-3-yl]To a solution of methyl ethyl } pyridine-2-carboxylate (1.80g,5.78mmol) in THF (28.9mL) was added a solution of LiOH (415mg,17.3mmol) in water (5.8 mL). After 2hr, the reaction mixture was diluted with DCM and 1.0 MHCl. NaCl was added to saturation and then saturated NaHCO was added slowly at RT3Aqueous solution until the mixture reaches pH 2. The layers were separated and washed with CHCl3The aqueous layer was extracted 5 times with 2-propanol (3: 1). The organic layers were combined and washed with Na2SO4Drying, filtration and concentration under reduced pressure gave the title compound (1.71g, 99% yield). LC-ES/MS (m/z): 298.0(M + H).
Preparation example 10
5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxylic acid
Figure GDA0002319866120000281
To 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy at RT]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxylic acid methyl ester (16.25g, 36.8mmol) in MeOH (162mL) and water (32.5mL) LiOH (2.64g, 110.0mmol) was added. The reaction mixture was stirred at RT for 1hr and 1.0M HCl (115mL,115.0 mmol) was added. The mixture was diluted with EtOAc and water and the layers were separated. The aqueous layer was extracted with EtOAc. The organic layers were combined and washed with Na2SO4Drying, filtration, concentration under reduced pressure, vacuum drying gave the title compound (16.2g, quantitative yield), which was suitable for use without further purification. LC-ES/MS (m/z): 428.2(M + H).
Preparation example 11
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Figure GDA0002319866120000282
To a stirred solution of 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxylic acid (16.2g,37.9mmol) and 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride (7.20g,41.7mmol) in DMF (162mL) was added diisopropylethylamine (39.6mL,227.0mmol) and BOP (19.4g,41.7mmol) over 10min at RT in three portions. The reaction mixture was stirred at RT for 1.5hr and 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride (1.4g,8.1 mmol) was added. The reaction mixture was stirred at RT for 45min and BOP (3.6g,8.1mmol) was added. The reaction mixture was stirred at RT for 1hr, diluted with MTBE and water. The layers were separated and the aqueous layer was extracted with MTBE. The combined organic layers were washed 2 times with water, filtered, concentrated under reduced pressure, and dried in vacuo to give the title compound (15g, 72% yield). LC-ES/MS (m/z): 546.2(M + H).
Preparation example 12
2, 6-dimethylpyridine-4-carbonitrile
Figure GDA0002319866120000291
Scheme 4, step a: to a stirred mixture of 4-bromo-2, 6-lutidine (5.09g, 26.5mmol) and DMF (40mL) under nitrogen at RT was added zinc cyanide (3.82g,31.9 mmol). Nitrogen was bubbled through the stirred suspension for 15min, and tetrakis (triphenylphosphine) palladium (0) (1.54g,1.33 mmol) was added. After heating the reaction mixture at 120 ℃ for 5.5hr, the mixture was cooled to RT and diluted with EtOAc (150 mL). The solids were removed by paper filtration and the filter cake was washed with EtOAc (50 mL). With 15% NH in sequence3The combined organic filtrate and washings were washed with aqueous solution (2X 50mL), water (50mL) and saturated aqueous NaCl solution, over Na2SO4Drying, filtering and concentrating under reduced pressure to obtain yellow solid. The crude product was purified by flash chromatography on silica eluting with hexane/ethyl acetate (gradient 9:1-1: 1). The pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (2.79g, 77% yield).1H NMR (CDCl3):δ2.61(s,6H),7.21(s,2H).
Alternative method for preparation 12
2, 6-dimethylpyridine-4-carbonitrile
Scheme 4, step a: equipped with a mechanical stirrer, a reflux condenser and N24-bromo-2, 6-lutidine (235.0g,1263.1mmol) was dissolved in anhydrous DMF (250 mL) in an inlet 3-neck round-bottom flask and N was added2Bubbling through the solution for 20 min. In N2Part of the solution (-150 mL) was transferred through cannula to another funnel. Zinc cyanide (150.0g,1277.4mmol) and tetrakis (triphenylphosphine) palladium (0) (15.0g,13.0mmol) were added to the reaction mixture by allowing N to react2The mixture was bubbled through for 15min to purge. The reaction mixture was heated to 90 ℃. 4-bromo-2, 6-lutidine in DMF was added dropwise over 30min with continued heating overnight. The mixture was cooled to RT, MTBE (. about.2L) was added, then water (1.5L) and 30% NH were added4Aqueous OH (800 mL); mixing the obtained mixtureThe mixture was stirred at RT for 30 min. The layers were separated and the aqueous layer was extracted 1 time with MTBE (. about.2L); the organic phases were combined with 10% NH4OH aqueous solution (2L) 1 time, Na2SO4Drying, filtration, and evaporation of the filtrate under reduced pressure gave the crude title compound (153g, 91.7% yield) as a pale yellow solid contaminated with-10% triphenylphosphine by-product, which was suitable for use without additional purification.1H NMR(CDCl3):δ2.61(s,6H),7.21(s,2H).
Preparation example 13
1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride
Figure GDA0002319866120000301
Scheme 4, step B: a solution of 2, 6-lutidine-4-carbonitrile (2.26g,16.7mmol) in EtOH (40mL) was added to a suspension of 10% Pd on carbon (405mg), EtOH (10mL) and concentrated aqueous HCl (6.9 mL). The reaction vessel was evacuated, purged with nitrogen, and H was introduced2(55 psi) while stirring the subsequent reaction mixture at RT for 16 hr. The reaction mixture was filtered through celite. The filter cake was washed with MeOH and the combined filtrate/washings were concentrated to give a yellow solid. The crude material was triturated with boiling 30% EtOH/EtOAc, cooled to RT and collected by filtration to give the title compound (2.64g, 75% yield). LC-ES/MS (m/z): 137.0(M + H).
Alternative method for preparation 13
1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride
Scheme 4, step B: the following procedure was carried out in two batches, which were combined after the complete hydrogenation reaction: 2, 6-lutidine-4-carbonitrile (77.39g,527.0mmol) was added to a solution containing 10% Pd/C (45.8g) in MeOH (800mL) and 4M HCl bis (HCl) equipped with a mechanical stirrer
Figure GDA0002319866120000302
A mixture of alkane solutions (500 mL) was placed in a 2L Parr autoclave. Sealing the autoclave with N2Followed by H2Thoroughly purifying the resulting mixtureSubstance, with H2Pressurize to 60psi while stirring at RT overnight. The reaction mixture was filtered and the filtrate was evaporated under reduced pressure. MeOH (. about.250 mL) was added to the resulting residue, stirred for 15hr, and MTBE (2.5L) was added slowly. The mixture was stirred at RT for 1hr, filtered and the solid was washed with MTBE (1L). The solid was dried under vacuum at RT overnight to give the title compound as a pale yellow solid (217.0g, 91.6% yield, combined two runs), which was suitable for use without additional purification. LC-ES/MS (m/z): 137.2(M + H), 92.5% purity, with 7.5% triphenylphosphine impurity present (1.57min, M/z: 263.0).
Preparation example 14
(3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one and
(3S) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one
Figure GDA0002319866120000311
Scheme 5, step a: equipped with a mechanical stirrer, a reflux condenser and N2N was passed through an inlet 3-neck round-bottom flask over a period of about 30min2Bubbling 3-methyl-1, 3-dihydropyrrolo [2,3-b ]]Pyridin-2-one (109g,735.69mmol) and [ (1S) -methanesulfonic acid 1- (6-bromo-3-pyridinyl) ethyl]A solution of the ester (207g, 738.92mmol) in dry DMF (1100mL) was purified. Cooling the obtained solution to 0 ℃, and adding Cs in portions2CO3(240g,736.59 mmol). Then by making N2The mixture was purged by bubbling for 15min and stirred at about 0 ℃ for about 40 hr. The reaction mixture was diluted with water (4L) and MTBE (3L). The organic extracts were washed successively with water and 5% aqueous LiCl, Na2SO4Drying, filtration, and concentration under reduced pressure gave the title compound as a diastereomer mixture in a 5:1 ratio (183.2g, 75% yield). LC-ES/MS (m/z,79Br/81Br):332.0,334.0(M+H).
preparation example 15
(3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one, (7, 7-dimethyl-2-oxo-norbornan-1-yl) methanesulfonate; (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one, (1S) - (+) -10-camphorsulfonic acid salt
Figure GDA0002319866120000321
Scheme 5, step B: equipped with a mechanical stirrer, a reflux condenser and N2To an inlet 3-neck round-bottom flask was added (3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one and (3S) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]A solution of diastereomeric mixture of pyridin-2-one (183.2g,553.5mmol) in EtOAc (1.3L) was heated to 50 ℃. (1S) - (+) -10-Camphorsulfonic acid (171.0g,736.1mmol) was added and the reaction mixture was heated at reflux for 30min, then cooled to RT and stirred overnight. The reaction mixture was filtered, the filter cake was washed with EtOAc, and the resulting white solid was dried in vacuo to give the title compound (288.3g, 92.5% yield). LC-ES/MS (m/z,79Br/81br): 332.0,334.0(M + H). from saturated NaHCO3Chiral HPLC of EtOAc-extracted aliquots of suspensions in aqueous solution: (
Figure GDA0002319866120000322
AD-H,4.6X 150mm, 100% EtOH with 0.2% DMEA, 1 mL/min, run 12min,254 nm): t is tR=7.66min,>96%ee.
Preparation example 16
(3R) -3- [ (1R) -1- (6-bromo-3-pyridinyl) ethyl ] -3-methyl-1H-pyrrolo [2,3-b ] pyridin-2-one
Figure GDA0002319866120000323
Scheme 5, step C: reacting (3R) -3- [ (1R) -1- (6-bromo-3-pyridyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one, (1S) - (+) -10-Camphorsulfonate (288.3g,512.01mmol) in NaHCO3Aqueous solution (371g,4416.4mmol,6L H)2O) and 5L EtOAc. The resulting biphasic mixture was stirred at RT for about 45min and separatedThe layers were combined and the aqueous phase was extracted with EtOAc (4L). The combined EtOAc phases were washed with saturated aqueous NaCl solution and Na2SO4Drying, filtering, and concentrating the filtrate under reduced pressure. The resulting residue was crystallized from MeOH (-1.7L) and the resulting solid was collected by filtration to give the title compound (244.4g, 59.7% yield) as a white crystalline solid. LC-ES/MS (m/z,79Br/81br): 332.0,334.0(M + H). Chiral HPLC (
Figure GDA0002319866120000324
AD-H,4.6X 150mm, 100% EtOH with 0.2% DMEA, 1 mL/min, run 12min,254 nm): t is tR=7.66min,>98%ee.
Preparation example 17
2-ethyl-6-methylpyridine-4-carbonitrile
Figure GDA0002319866120000331
Scheme 8, step a: ethyl magnesium bromide (18ml,17.7mmol) was added in portions to 2-chloro-6-methylisonicotonitrile (Bioorganic)&Medicinal Chemistry Letters,20(2), 576-580; 2010) (1.5g,9.8mmol), 1-methyl-2-pyrrolidone (10mL), THF (10mL), and iron (III) acetoacetate (521mg,1.47mmol) while stirring under nitrogen at RT. The reaction mixture was concentrated to remove most of the THF and quenched with water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed successively with water and saturated aqueous NaCl and Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a crude product. The crude product was purified by flash chromatography on silica, eluting with 15% hexane/ethyl acetate. Pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (0.53g, 37%). LC-ES/MS (m/z): 147.2(M + H).
Preparation example 18
1- (2-ethyl-6-methylpyridin-4-yl) methylamine dihydrochloride
Figure GDA0002319866120000332
Scheme 8, step B:2-Ethyl-6-methylpyridine-4-carbonitrile (0.37g,2.5mmol) in 2M NH3A solution of (2mol/l) in MeOH (12.5mL) was added to Raney nickel (0.5g) in 2M NH3To a suspension in MeOH solution (12.5 mL). Purging the reaction vessel with nitrogen and introducing H2(60psi) while shaking the subsequent reaction mixture at 40 ℃ for 15 minutes. Reuse H2The reaction mixture was pressurized (60psi) and shaken continuously for 4 hr. The reaction mixture was filtered. The crude material was diluted with excess 3N HCl in MeOH and concentrated to give a green oil. The crude oil was triturated and concentrated sequentially with the following solvents: toluene, acetonitrile, methanol/toluene and acetonitrile/toluene, removal of the solvent gave the title compound as a green solid. LC-ES/MS (m/z): 151.0(M + H).
Preparation example 19
2-cyclopropyl-6-methyl-pyridine-4-carbonitrile
Figure GDA0002319866120000341
Scheme 8, step a: 2-chloro-6-methyl-pyridine-4-carbonitrile (j.med.chem.,59(1) 313-Fluorocarboxylic acid, 327,2016,2.0g,12.7mmol), cyclopropylboronic acid (1.84g,20.3mmol) and K3PO4(5.56g,25.4mmol) was slurried in a mixture of toluene (40mL) and water (2 mL). Addition of Pd (OAc)2(291mg,1.27mmol) and tricyclohexylphosphine tetrafluoroborate (946mg,2.554 mmol), and the reaction mixture was heated at 110 ℃ for 16hr under a nitrogen balloon. The reaction mixture was cooled to RT, diluted with EtOAc (50mL) and filtered through a celite bed. The filtrate was washed with saturated aqueous NaCl solution and Na2SO4Dried, filtered and concentrated under reduced pressure to give an amber oil. The resulting residue was purified by flash chromatography on silica, eluting with a gradient of 2-20% EtOAc in hexanes over 30min, to give the title compound (1.66g, 82% yield) as a light yellow solid after evaporation of the solvent.1H NMR(CDCl3):δ1.05-1.06(m,4H)2.03-2.09(m,1H),2.54(s,3H),7.11 (s,1H),7.14(s,1H).
Preparation example 20
(2-cyclopropyl-6-methyl-4-pyridyl) methylamine dihydrochloride
Figure GDA0002319866120000342
Scheme 8, step B: 2-cyclopropyl-6-methyl-pyridine-4-carbonitrile (2.52g,15.9mmol) was reduced in a similar manner to that described in preparation 18 to give the title compound (3.57g, 95% yield). LC-ES/MS (m/z): 163.0(M + H).
Preparation example 21
2-isopropyl-6-methyl-pyridine-4-carboxylic acid methyl ester
Figure GDA0002319866120000343
A solution of isopropylmagnesium chloride in THF (2.0M,7.53mL,15.1mmol) was added dropwise to methyl 2-chloro-6-methyl-pyridine-4-carboxylate (1.92g,10.0mmol), MnCl, in an ice/water bath under nitrogen over 8 minutes while stirring2(0.065g,0.502mmol) and THF (25 mL). Stirring in cold bath for 4hr, adding saturated NH4The reaction was quenched with aqueous Cl and extracted with EtOAc (2X 50 mL). The combined extracts were washed with saturated aqueous NaCl solution and Na2SO4Dried, filtered and concentrated under reduced pressure to give an amber oil. The crude product was purified by flash chromatography on silica eluting with hexane/ethyl acetate (gradient 50:1-2: 1). The pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (0.683g, 35% yield).1HNMR(CDCl3):δ1.34(d,J=6.9Hz, 6H),2.63(s,3H),3.12-3.19(m,1H),3.96(s,3H),7.54(s,1H),7.56(s,1H).
Preparation example 22
2- [ (2-isopropyl-6-methyl-4-pyridyl) methyl ] isoindoline-1, 3-dione
Figure GDA0002319866120000351
Sodium borohydride (0.231g,6.01mmol) was added to a solution of methyl 2-isopropyl-6-methyl-pyridine-4-carboxylate (0.683g,3.53mmol) in EtOH (15mL) at RT. After stirring overnight, the solvent was removed under reduced pressureThe remaining oil was diluted with saturated aqueous NaCl and extracted with EtOAc (2X 50 mL). With Na2SO4The combined extracts were dried, filtered, and concentrated under reduced pressure to give 652mg of crude (2-isopropyl-6-methyl-4-pyridyl) methanol as an amber solid. The crude alcohol was dissolved in DCM (20mL) and treated with thionyl chloride (0.514mL,7.02 mmol). After stirring at RT for 4hr, the reaction mixture was concentrated under reduced pressure; dissolve in toluene and concentrate (2 ×). The crude alkyl chloride was dissolved in DMF (10mL) and potassium phthalimide (1.32g,7.02mmol) was added. The suspension was stirred at RT for 3.5hr and diluted with water (100 mL). The resulting suspension was stirred at RT for 1 hour and the solid was collected by filtration. The crude product was purified by flash chromatography on silica eluting with DCM/ethyl acetate (gradient 50:1-4: 1). The pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (0.332g, 32% yield).1H NMR (CDCl3):δ1.29(d,J=6.9Hz,6H),2.51(s,3H),3.06-3.08(m,1H),4.80(s, 2H),6.96(s,1H),7.00(s,1H),7.76-7.79(m,2H),7.89-7.92(m,2H).
Preparation example 23
(2-isopropyl-6-methyl-4-pyridyl) methylamine
Figure GDA0002319866120000361
Hydrazine monohydrate (0.70mL,1.41mmol) was added to 2- [ (2-isopropyl-6-methyl-4-pyridyl) methyl at RT]Isoindoline-1, 3-dione (0.332g,1.13mmol) and EtOH (10 mL). After refluxing for 1.5hr, the reaction mixture was cooled to RT and the solids were removed by paper filtration. The filter cake was washed with EtOH (10mL) and the combined filtrate/washings were concentrated under reduced pressure to give the title compound (0.179g, 96% yield).1H NMR(CDCl3):δ1.31(d,J=6.8Hz,6H),1.56-1.63(bs,2H),2.54(s,3H),3.02-3.09(m,1H),3.86(s,2H),6.95(s,2H).
Preparation example 24
2-cyclobutyl-6-methyl-pyridine-4-carbonitrile
Figure GDA0002319866120000362
Scheme 8, step a: cyclobutylzinc bromide (0.5M in THF, 3.28mmol) was added dropwise to degassed 2-chloro-6-methyl-pyridine-4-carbonitrile (1.64 mmol) and [1,1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (0.164mmol) in 1, 4-bis
Figure GDA0002319866120000364
In an alkane. Heating to 80 deg.C for 1hr, and then cooling to RT. Water (5mL) was added and stirred rapidly for 5 minutes. The solids were removed by filtration through celite. Washed with EtOAc and the layers separated. The organic layer was washed with water and saturated aqueous NaCl solution. With Na2SO4Drying, filtering, and concentrating under reduced pressure. The resulting residue was purified by flash chromatography on silica, eluting with a gradient of 10-30% EtOAc in heptane, to give the title compound (232mg, 82%) after evaporation of the solvent. LC-ES/MS (m/z): 173.0(M + H).
Preparation example 25
(2-cyclobutyl-6-methyl-4-pyridyl) methylamine dihydrochloride
Figure GDA0002319866120000363
Scheme 8, step B: the title compound was prepared essentially by the method described in preparation 18 using 2-cyclobutyl-6-methyl-pyridine-4-carbonitrile. LC-ES/MS (m/z): 177.0(M + H).
Preparation example 26
2-cyclopentyl-6-methyl-pyridine-4-carbonitrile
Figure GDA0002319866120000371
Scheme 8, step a: the title compound was prepared essentially by the method described in preparation 24 using cyclopentylzinc bromide.1H NMR(CDCl3):δ1.62-1.78(m,4H),1.78-1.89(m, 2H),2.00-2.14(m,2H),2.57(s,3H),3.09-3.25(m,1H),7.16(s,1H),7.19(s, 1H).
Preparation example 27
(2-cyclopentyl-6-methyl-4-pyridyl) methylamine
Figure GDA0002319866120000372
Scheme 8, step B: the title compound is prepared essentially by the method described in preparation 18 using 2-cyclopentyl-6-methyl-pyridine-4-carbonitrile. LC-ES/MS (m/z): 191.0(M + H).
Preparation example 28
4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] pyrimidin-6-one
Figure GDA0002319866120000373
Scheme 7, step a: to the solution of 4-chloro-7- (2, 4-dimethoxybenzyl) -5, 7-dihydro-6H-pyrrolo [2,3-d]To a solution of pyrimidin-6-one (US 2010/0120801,6.02g,18.8mmol) in DMF (37.7mL) was added NaH (60% in mineral oil, 753mg,18.8mmol) slowly. The reaction mixture was stirred at RT for 30 min. Dropwise adding CH3I (1.17mL,18.8mmol) and the reaction mixture was stirred at RT for 30 min. Adding saturated NH4Aqueous Cl and EtOAc, and the layers were separated. The organic layer was washed with brine, Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a solid residue. The residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 9:1-1:1) to give, after evaporation of the solvent, the title compound (1.30g, 20%). LC-ES/MS (m/z)35Cl/37Cl):334.0/336.0(M+H).
Preparation example 29
4- [ (1S) -1-hydroxyethyl ] benzoic acid methyl ester
Figure GDA0002319866120000381
Palladium (II) acetate (335mg,1.49mmol), 1' -bis (diphenylphosphino) ferrocene (993mg,1.79mmol), (1S) -1- (4-bromophenyl) ethanol (3.0g,14.9mmol), anhydrous ACN (100mL), anhydrous MeOH (70mL), and TEA (5.2mL,37mmol) were combined in a 300mL Parr autoclave equipped with a mechanical stirrer. Sealing the autoclave with CO2Purification with CO2Pressurized to 689kPa and heated to 100 ℃ while stirring. After 6hr at 100 ℃, the reaction mixture was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure to give a solid residue. The residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 9:1-1:1) to give, after evaporation of the solvent, the title compound (2.7g, 100%). LC-ES/MS (m/z): 181.2(M + H).
Preparation example 30
4- { (1S) -1- [ (methylsulfonyl) oxy ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000382
Stirring 4- [ (1S) -1-hydroxyethyl at 0 deg.C]To a solution of methyl benzoate (6.16g,34.2mmol) and TEA (7.15mL,51.3mmol) in DCM (51mL) was added methanesulfonyl chloride (3.17mL,41.0 mmol). After 1hr at 0 deg.C, water and DCM were added and the layers were separated. The aqueous layer was extracted with DCM. The organic layers were combined and successively saturated NaHCO3Aqueous solution and then washed with saturated NaCl; with Na2SO4Drying; filtration and concentration under reduced pressure gave the title compound (8.71g, 99% yield).1HNMR(CDCl3)δ1.73(d,J=6.7Hz,3H),2.80(s,3H),3.93(s,3H), 5.77(q,J=6.7Hz,1H),7.47-7.49(m,2H),8.06-8.09(m,2H).
Preparation example 31
4- { (1R) -1- [ (5R) -4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000391
Scheme 7, step B (X ═ CH): stirring 4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-5, 7-dihydro-6H-pyrrolo [2,3-d ] at 0 ℃ under nitrogen]Pyrimidin-6-one (3.68g,10.9mmol) and 4- { (1S) -1- [ (methylsulfonyl) oxy]To a solution of methyl ethyl } benzoate (2.82g,10.9mmol) in DMF (54.5mL) was added Cs2CO3(4.26g,13.1 mmol). Using nitrogen gas to fullyThe reaction mixture was purged and gradually warmed from 0 ℃ to RT over 19hr while stirring. DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The crude product was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 1:0-1: 1). The pure chromatographic fractions were combined and the solvent was evaporated to give the title compound (3.38g, 61% yield) as diastereomer in a 15:1 ratio. LC-ES/MS (m/z)35Cl/37Cl):496.0/498.0(M+H).
The impure chromatographic fractions were concentrated under reduced pressure and re-purified by flash chromatography on silica eluting with DCM/EtOAc (gradient 1:0-9:1) to give, after evaporation of the solvent, a further title compound (991mg, 18%) as diastereomer in a 5:1 ratio. LC-ES/MS (m/z)35Cl/37Cl):496.0/498.0 (M+H).
Preparation example 32
4- { (1R) -1- [ (5R) -7- (2, 4-Dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000401
Scheme 7, step C (X ═ CH): palladium (5% on carbon, 24mg,0.23mmol), 4- { (1R) -1- [ (5R) -4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2, 3-d)]Pyrimidin-5-yl]Ethyl } benzoic acid methyl ester (0.2g,0.4mmol,15:1dr), anhydrous MeOH (10mL), and TEA (0.2mL,1mmol) were combined in a 70mL Parr shake flask. Sealing the Parr flask with N2Purification with H2Purification with H2The pressure was increased to 138 kPa. The reaction mixture was shaken at RT for 70min and then filtered.
Palladium (294 mg,2.76mmol on 5% carbon), 4- { (1R) -1- [ (5R) -4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2, 3-d)]Pyrimidin-5-yl]Ethyl } benzoic acid methyl ester (3.1g,6.0mmol,15:1dr), anhydrous MeOH (150mL), and TEA (2.0mL, 14.3mmol) were combined in a 500mL Parr shake flask. Sealing the Parr flask with N2Purification with H2Purification with H2The pressure was increased to 138 kPa. The reaction mixture was shaken at RT for 80min and then filtered. The two filtrates from the hydrodechlorination reaction were combined and concentrated under reduced pressure to give a residue.
Palladium (5% on carbon, 0.10g,0.94mmol), 4- { (1R) -1- [ (5R) -4-chloro-7- (2, 4-dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2, 3-d)]Pyrimidin-5-yl]Ethyl } benzoic acid methyl ester (991mg,1.93mmol,5:1dr), anhydrous MeOH (50mL), and TEA (0.70mL, 5.0mmol) were combined in a 500mL Parr shake flask. Sealing the Parr flask with N2Purification with H2Purification with H2The pressure was increased to 138 kPa. The reaction mixture was shaken at RT for 85min and then filtered. The filtrate was concentrated under reduced pressure to give a residue.
The entire residue from the hydrodechlorination reaction described above was dissolved in DCM and combined. Adding saturated NaHCO3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a solid residue. The residue was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 4:1-0: 1). The impure chromatographic fractions were concentrated under reduced pressure and re-purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 3:1-0: 1). All pure chromatographic fractions were combined, concentrated under reduced pressure and the solvent evaporated to give the title compound (2.99g, 74% yield) as a single diastereomer. LC-ES/MS (m/z): 462.2(M + H).1H NMR(CDCl3)δ1.18 (d,J=7.1Hz,3H),1.39(s,3H),3.35(q,J=7.1Hz,1H),3.74(s,3H),3.79(s, 3H),3.92(s,3H),4.87(d,J=14.9Hz,1H),4.92(d,J=14.9Hz,1H),6.38(dd, J=2.4,8.4Hz,1H),6.41(d,J=2.4Hz,1H),7.01(d,J=8.4Hz,1H), 7.12-7.17(m,2H),7.88-7.92(m,2H),7.94(s,1H),8.78(s,1H).
Preparation example 33
4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000411
Scheme 7, step D (X ═ CH): 4- { (1R) -1- [ (5R) -7- (2, 4-Dimethoxybenzyl) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] is reacted at RT]Pyrimidin-5-yl]A solution of methyl ethyl } benzoate (1.50g,3.25mmol) in anisole (1.77mL,16.3mmol) and TFA (4.92mL,65.0 mmol) was divided into 5 aliquots, each of which was placed into a 4mL vial. The vial was tightly closed and heated to 120 ℃ while stirring. After 6hr, the reaction mixture was cooled to RT. The contents of all vials were poured into DCM and saturated NaHCO3In the mixture of aqueous solutions, the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The residue was purified by flash chromatography on silica eluting with DCM/EtOAc (gradient 1:0-0:1) to give the title compound (495mg, 49% yield) after evaporation of the solvent. LC-ES/MS (m/z): 312.0(M + H).
Preparation example 34
4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzoic acid
Figure GDA0002319866120000412
Scheme 7, step E (X ═ CH): to 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] at RT]Pyridin-3-yl]To a solution of methyl ethyl } pyridine-2-carboxylate (480mg,1.54 mmol) in THF (7.7mL) was added a solution of LiOH (111mg,4.63mmol) in water (1.54 mL). After 18hr, the reaction mixture was diluted with DCM and saturated aqueous NaCl solution. 1.0M HCl was added slowly at RT until the mixture reached pH 2. The layers were separated and washed with CHCl3The aqueous layer was extracted 3 times with 2-propanol (3: 1). The organic layers were combined and washed with Na2SO4Drying, filtration, and concentration under reduced pressure gave the title compound (449mg, 98% yield). LC-ES/MS (m/z): 298.0(M + H).
Preparation example 35
1- (2, 6-dimethylpyridin-4-yl) methylamine
Figure GDA0002319866120000421
2, 6-lutidine-4-carbonitrile (2.0g,15.1mmol) was dissolved in 2.0M NH in a 500mL Parr flask3To a solution of Raney nickel (0.5g,9mmol) in 2.0M NH3To a suspension in MeOH solution (75 mL). Sealing the Parr flask with N2Purification with H2Purification with H2The pressure was increased to 414 kPa. The reaction mixture was heated to 40 ℃ and allowed to cool slowly to RT while shaking for 18 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a green residue. The residue was purified by flash chromatography on silica eluting with hexane/DCM/IPAm (gradient 9:0:1-6:3: 1). The chromatographic fractions containing the product were concentrated under reduced pressure and re-purified by flash chromatography on silica, eluting with hexane/DCM/IPAm (45:45:10) to give the title compound (1.02g, 49% yield) after evaporation of the solvent. LC-ES/MS (m/z): 137.0(M + H).
Preparation example 36
5- [ (1S) -1-hydroxyethyl ] pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000422
Palladium (II) acetate (180mg,0.762mmol), 1' -bis (diphenylphosphino) ferrocene (535 mg,0.936mmol), (1S) -1- (6-bromo-3-pyridinyl) ethanol (1.6g,7.9mmol), anhydrous MeOH (40mL), anhydrous ACN (60mL), and TEA (2.8mL,20mmol) were combined in a 300mL Parr autoclave equipped with a mechanical stirrer. The autoclave was sealed, purged with CO, pressurized to 689kPa with CO, and heated to 85 ℃ while stirring. After 2hr, the reaction mixture was cooled to RT and filtered. The filtrate was concentrated under reduced pressure to give an oil. The residue was purified by flash chromatography on silica, eluting with hexanes/EtOAc, to give the title compound (1.5g, 100% yield). LC-ES/MS (m/z): 182.0 (M + H).
Preparation example 37
5- [ (1S) -1-Methylsulfonyloxyethyl ] pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000431
Reacting 5- [ (1S) -1-hydroxyethyl at 0 DEG C]Pyridine-2-carboxylic acid methyl ester (1.5g,8.3mmol) was stirred in DCM (20mL) with TEA (1.7mL,12 mmol). Methanesulfonyl chloride (0.78mL,9.9 mmol) was added dropwise and stirred for 2 hr. The solution was diluted with DCM and NaHCO3And saturated aqueous NaCl solution, followed by Na2SO4Drying, filtration, and concentration gave the title compound (2.1g, 100% yield).1H NMR(CDCl3)δ1.77(d,J=6.7Hz,3H),2.93(s,3H),4.03(s,3H),5.85(q, J=6.7Hz,1H),7.91(dd,J=2.2,8.1Hz,1H),8.18(d,J=8.1Hz,1H),8.76(d, J=2.2Hz,1H).
Preparation example 38
5- [ (1R) -1- [ (5R) -4-chloro-7- [ (2, 4-dimethoxyphenyl) methyl ] -5-methyl-6-oxo-pyrrolo [2,3-c ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000432
Scheme 7, step B (X ═ N): 4-chloro-7- [ (2, 4-dimethoxyphenyl) methyl]-5-methyl-5H-pyrrolo [2,3-c]Pyridazin-6-one (2.0g,6.0mmol) and 5- [ (1S) -1-methylsulfonyloxyethyl]Pyridine-2-carboxylic acid methyl ester (1.86g,7.2mmol) was combined in DMF (50ml), cooled to 0 ℃ and purged thoroughly with nitrogen. Cesium carbonate (2.35g,1.2mmol) was added and the mixture was purged thoroughly with nitrogen and stirred at 0 ℃ overnight. The reaction solution was diluted with EtOAc, washed with water and saturated aqueous NaCl solution. With Na2SO4The organic layer was dried, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography on silica, eluting with hexanes/EtOAc, to give the title compound (2.3g, 77% yield). LC-ES/MS (m/z): 497.0 (M + H).
Preparation example 39
5- [ (1R) -1- [ (5R) -7- [ (2, 4-Dimethoxyphenyl) methyl ] -5-methyl-6-oxo-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000441
Scheme 7, step C (X ═ N): palladium (5% on carbon, 240mg,2.4mmol) was charged to a 500mL Parr autoclave, purged with nitrogen. Methyl 5- [ (1R) -1- [ (5R) -4-chloro-7- [ (2, 4-dimethoxyphenyl) methyl ] -5-methyl-6-oxo-pyrrolo [2,3-c ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylate (2.2 g,4.4mmol) and MeOH (100mL) were added. The autoclave was sealed, purged with nitrogen and then with hydrogen. The autoclave was pressurized to 414kPa with hydrogen and shaken at RT for 4 hr. The reaction solution was filtered and concentrated under reduced pressure to give the title compound (2.1g, 100% yield). LC-ES/MS (m/z): 463.2(M + H).
Preparation example 40
5- [ (1R) -1- [ (5R) -5-methyl-6-oxo-7H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylic acid methyl ester
Figure GDA0002319866120000442
Scheme 7, step D (X ═ N): reacting 5- [ (1R) -1- [ (5R) -7- [ (2, 4-dimethoxyphenyl) methyl]-5-methyl-6-oxo-pyrrolo [2,3-d]Pyrimidin-5-yl]Ethyl radical]Pyridine-2-carboxylic acid methyl ester (750mg,1.62 mmol) was combined in a microwave vial with anisole (2.5mL,23mmol) and trifluoroacetic acid (2.5mL,32 mmol). The vial was capped and heated in a microwave at 140 ℃ for 3 hr. By CH2Cl2The solution was diluted with NaHCO3And (6) washing. With Na2SO4The organic layer was dried, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography on silica gel using CH2Cl2EtOAc/MeOH elution provided the title compound (250mg, 49% yield). LC-ES/MS (m/z): 313.0(M + H).
Preparation example 41
5- [ (1R) -1- [ (5R) -5-methyl-6-oxo-7H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylic acid
Figure GDA0002319866120000451
Scheme 7, step E (X ═ N): at RT to 5- [ (1R) -1- [ (5R) -5-methyl-6-oxo-7H-pyrrolo [2,3-d]Pyrimidin-5-yl]Ethyl radical]Pyridine-2-carboxylic acid methyl ester(250mg,0.80mmol) to a solution in THF (5mL) and water (1mL) was added LiOH (58mg,2.4 mmol). The reaction mixture was stirred at RT for 48 hr. The mixture was diluted with DCM (10mL) and 1M HCl was added to reach pH-3. Adding saturated NaCl solution, and adding CHCl3The mixture was extracted 3 times with/iPrOH (3: 1). The organic layers were combined and washed with Na2SO4Drying, filtration, and concentration under reduced pressure gave the title compound (188mg, 79% yield). LC-ES/MS (m/z): 297.0(M + H).
Preparation example 42
2-isopropyl-6-methyl-pyridine-4-carboxylic acid methyl ester
Figure GDA0002319866120000452
A2M solution of isopropylmagnesium chloride in THF (7.53mL, 15.1mmol) was added dropwise to methyl 2-chloro-6-methyl-pyridine-4-carboxylate (1.92g,10.0mmol), MnCl, under nitrogen in an ice/water bath over 8 minutes2(65mg,0.5mmol) and THF (25mL) while stirring. After stirring for 4 hours in a cold bath, saturated NH was used4The reaction was quenched with aqueous Cl and extracted with EtOAc (2X 50 mL). The combined extracts were washed with saturated aqueous NaCl solution and Na2SO4Dried, filtered and concentrated under reduced pressure to give an amber oil. The crude product was purified by flash chromatography on silica eluting with hexane/ethyl acetate (gradient 50:1-2: 1). The pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (0.683g, 35% yield).1HNMR(CDCl3):δ1.34(d,J=6.9Hz, 6H)2.63(s,3H),3.12-3.19(m,1H),3.96(s,3H),7.54(s,1H),7.56(s,1H).
Preparation example 43
2- [ (2-isopropyl-6-methyl-4-pyridyl) methyl ] isoindoline-1, 3-dione
Figure GDA0002319866120000461
Sodium borohydride (231mg,6.0mmol) was added to a solution of methyl 2-isopropyl-6-methyl-pyridine-4-carboxylate (683mg,3.53mmol) in EtOH (15mL) at RT. Stirring the mixtureAfter overnight, the solvent was removed under reduced pressure, the remaining oil was diluted with saturated aqueous NaCl and extracted with EtOAc (2X 50 mL). With Na2SO4The combined extracts were dried, filtered, and concentrated under reduced pressure to give 652mg of crude (2-isopropyl-6-methyl-4-pyridyl) methanol as an amber solid. The crude alcohol was dissolved in DCM (20mL) and treated with thionyl chloride (0.51mL,7.02 mmol). After stirring at RT for 4hr, the reaction mixture was concentrated under reduced pressure, dissolved in toluene, and concentrated (2 ×). The crude alkyl chloride was dissolved in DMF (10mL) and potassium phthalimide (1.32g,7.02mmol) was added. The suspension was stirred at RT for 3.5h and diluted with water (100 mL). The resulting suspension was stirred at RT for 1hr and the solid was collected by filtration. The crude product was purified by flash chromatography on silica eluting with DCM/ethyl acetate (gradient 50:1-4: 1). The pure chromatographic fractions were combined and concentrated under reduced pressure to give the title compound (332mg, 32% yield).1H NMR(CDCl3):δ1.29(d,J=6.9Hz,6H)2.51(s,3H),3.06-3.08(m,1H),4.80 (s,2H),6.96(s,1H),7.00(s,1H),7.76-7.79(m,2H),7.89-7.92(m,2H).
Preparation example 44
(2-isopropyl-6-methyl-4-pyridyl) methylamine
Figure GDA0002319866120000462
Hydrazine monohydrate (0.7mL,1.41mmol) was added to 2- [ (2-isopropyl-6-methyl-4-pyridyl) methyl at RT]Isoindoline-1, 3-dione (332mg,1.13mmol) and EtOH (10 mL). After 1.5 hours at reflux, the reaction mixture was cooled to RT and the solids were removed by paper filtration. The filter cake was washed with EtOH (10mL) and the combined filtrate/washings were concentrated under reduced pressure to give the title compound (0.179g, 96% yield).1H NMR(CDCl3):δ1.31(d,J=6.8Hz, 6H),1.56-1.63(br-s,2H),2.54(s,3H),3.02-3.09(m,1H),3.86(s,2H),6.95(s, 2H).
Preparation example 45
(6-chloro-4-methyl-2-pyridyl) methanol
Figure GDA0002319866120000471
Sodium borohydride (905mg,23.4mmol) was added in one portion to a solution of ethyl 6-chloro-4-methylpyridine-2-carboxylate (2.81g,13.8mmol) in EtOH (25 mL). The reaction mixture was stirred at RT for 18hr and concentrated under reduced pressure. The resulting residue was diluted with saturated aqueous NaCl, extracted 2 times with EtOAc and washed with Na2SO4The organic extract was dried, filtered, and concentrated under reduced pressure to give the title compound, which was suitable for use without additional purification. LC-ES/MS (m/z)35Cl/37Cl):158.0/160.0 (M+H).
Preparation example 46
2-chloro-6- (chloromethyl) -4-methyl-pyridine
Figure GDA0002319866120000472
To a solution of (6-chloro-4-methyl-2-pyridinyl) methanol (2.3g,13.8mmol) in DCM (25mL) under a nitrogen atmosphere was added thionyl chloride (2mL,27.5 mmol). The reaction mixture was stirred at RT for 4.5hr, concentrated under reduced pressure, the residue redissolved with toluene, concentrated under reduced pressure again for 2 times, and then dried in a vacuum oven at 45 ℃ overnight to give the title compound (2.36g, 97% yield) as a bright amber oil.1H NMR(CDCl3):δ2.37(s,3H),4.59(s,2H),7.11(s,1H),7.24(s,1H).
Preparation example 47
2- [ (6-chloro-4-methyl-2-pyridyl) methyl ] isoindoline-1, 3-dione
Figure GDA0002319866120000473
Potassium phthalimide (2.98g,15.8mmol) was added to a solution of 2-chloro-6- (chloromethyl) -4-methyl-pyridine (2.36g,13.1mmol) in DMF (25mL) under a stream of nitrogen at RT. Stirring was continued for 3.5hr and additional phthalimide (523mg,2.8mmol) was added over 72hr while stirring was continued at RT. The reaction mixture was diluted with water (150mL) and the mixture was stirred for 30 min. The resulting white precipitate was collected by vacuum filtration,the solid was dried in a vacuum oven at 35 ℃ overnight to give the title compound as a white solid (3.6g, 96% yield).1H NMR (CDCl3):δ2.29(s,3H),4.94(s,2H),6.92(s,1H),7.04(s,1H),7.73-7.79(m, 2H),7.88-7.93(m,2H).
Preparation example 48
6- [ (1, 3-dioxoisoindolin-2-yl) methyl ] -4-methyl-pyridine-2-carbonitrile
Figure GDA0002319866120000481
Passing nitrogen through 2- [ (6-chloro-4-methyl-2-pyridyl) methyl group]Isoindoline-1, 3-dione (1.3g,4.6mmol), Zinc cyanide (420mg,3.5mmol), [1,1' -bis (diphenylphosphino) ferrocene dichloride]A suspension of palladium (II) (174mg,0.23mmol) and elemental zinc (76mg,1.2mmol) in DMF (20mL) was bubbled for 10 min. The reaction mixture was heated in an oil bath at 120 ℃ for 5.5 hr. The mixture was cooled to RT, diluted with EtOAc (100mL) and filtered through paper to remove any insoluble material. With 15% NH in sequence4Washing the filtrate with OH aqueous solution, water and saturated NaCl aqueous solution; with Na2SO4The organic layer was dried, filtered and concentrated under reduced pressure. The resulting residue was purified by flash chromatography on silica eluting with 10-60% EtOAc in hexanes over 35min to give the title compound (1.04g, 80.5% yield) as a light yellow solid after evaporation of the solvent.1H NMR(CDCl3):δ2.39(s,3H),5.00 (s,2H),7.28(s,1H),7.40(s,1H),7.74-7.80(m,2H),7.88-7.94(m,2H).
Preparation example 49
6- (aminomethyl) -4-methyl-pyridine-2-carbonitrile dihydrochloride
Figure GDA0002319866120000482
Hydrazine hydrate (371 uL, 7.5mmol) was added to 6- [ (1, 3-dioxoisoindolin-2-yl) methyl]-4-methyl-pyridine-2-carbonitrile (1.04g,3.7mmol) in EtOH (20mL) and the resulting mixture was heated at reflux for 1.5 hr. Cooling the reaction mixture, filtering, and concentrating under reduced pressureAnd (4) condensing the filtrate. The resulting residue was dissolved in MeOH (. about.25 mL), loaded onto an SCX column (10g), purified with 1:1 MeOH: DCM (30mL), MeOH (20mL), and 2M NH3MeOH (50 mL). The methanolic ammonia fraction was concentrated under reduced pressure to give a pale yellow oil, which was dissolved in THF (15mL) and washed with 4N HCl in 1, 4-bis
Figure GDA0002319866120000493
Alkane solution (2.5 mL). The mixture was stirred at RT for 15min and the resulting solid was collected by vacuum filtration. The crystalline cake was dried in a vacuum oven at 45 ℃ overnight to give the title compound (541mg, 66% yield) as a pale yellow solid.1HNMR (DMSO-d6):δ2.41(s,3H),4.22(q,J=5.6Hz,2H),7.70(s,1H),7.95(s,1H), 8.55(br-s,2H).
Preparation example 50
(2-chloro-6-methylpyridin-4-yl) methanol
Figure GDA0002319866120000491
Prepared essentially by the method described in preparation 45 from methyl 2-chloro-6-methylpyridine-4-carboxylate (2.4g,12.5mmol) to give the title compound (2.0g, 97% yield).1H NMR (CDCl3):δ2.54(s,3H),4.71(s,2H),7.07(s,1H),7.16(s,1H).
Preparation example 51
2-chloro-4- (chloromethyl) -6-methylpyridine
Figure GDA0002319866120000492
Prepared essentially by the method described in preparation 46 from (2-chloro-6-methylpyridin-4-yl) methanol (1.97g,12.1mmol) to give the title compound (2.28g, 99.5% yield).1H NMR (DMSO-d6):δ2.45(s,3H),4.74(s,2H),7.33(s,1H),7.37(s,1H).
Preparation example 52
2- [ (2-chloro-6-methyl-4-pyridyl) methyl ] isoindoline-1, 3-dione
Figure GDA0002319866120000501
Prepared essentially by the method described in preparation 47 from 2-chloro-4- (chloromethyl) -6-methylpyridine (2.28g,12mmol) to give the title compound (3.67g, 98.7% yield).1H NMR (DMSO-d6):δ2.41(s,3H),4.77(s,2H),7.21(s,1H),7.30(s,1H),7.85-7.94 (m,4H).
Preparation example 53
4- [ (1, 3-dioxoisoindolin-2-yl) methyl ] -6-methyl-pyridine-2-carbonitrile
Figure GDA0002319866120000502
Prepared by essentially the method described in preparation 48 from 2- [ (2-chloro-6-methyl-4-pyridyl) methyl]Isoindoline-1, 3-dione (3.66g,11.9mmol) was prepared to give the title compound (1.7g, 51.5% yield).1H NMR(CDCl3):δ2.58(s,3H),4.84(s,2H),7.36(s,1H),7.51 (s,1H),7.75-7.81(m,2H),7.88-7.93(m,2H).
Preparation example 54
4- (aminomethyl) -6-methylpyridine-2-carbonitrile
Figure GDA0002319866120000503
Essentially by the method described in preparation 49 starting from 4- [ (1, 3-dioxoisoindolin-2-yl) methyl]-6-methyl-pyridine-2-carbonitrile (1.69g,6.1mmol) was prepared to give the title compound (379 mg, 41% yield).1HNMR(CDCl3):δ1.47(br s,2H),2.59(s,3H),3.94(s, 2H),7.36(s,1H),7.53(s,1H).
Preparation example 55
4- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] methyl } -2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000511
To 3-methyl-1- { [2- (trimethylsilyl) ethoxy ] solution stirred at 0 ℃ under nitrogen]Methyl } -1, 3-dihydro-2H-pyrrolo [2, 3-b)]Pyridin-2-one (8.53g,30.6mmol) and 4- { (1S) -1- [ (methylsulfonyl) oxy]Methyl Ethyl } benzoate (8.71g,33.7mmol) in DMF (153mL) was added Cs2CO3(12.0g,36.8 mmol). The reaction mixture was purged thoroughly with nitrogen, stirred at 0 ℃ for 43hr and gradually warmed to RT. DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering, and concentrating under reduced pressure to obtain a crude product, which is a diastereomer mixture at a ratio of-5: 1. The crude product was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 4:1-3: 2). The impure chromatographic fractions were concentrated under reduced pressure and re-purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 9:1-3: 2). The pure fractions from both chromatographic purifications were combined and the solvent was evaporated to give the title compound 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy-1- { []Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } benzoic acid methyl ester, isomer 1(10.16g, 75%). LC/MS (m/z): 441.2(M + H).1H NMR(CDCl3)δ-0.03(s,9H),0.91-1.03(m, 2H),1.18(d,J=7.1Hz,3H),1.33(s,3H),3.39(q,J=7.1Hz,1H),3.58-3.68 (m,2H),3.92(s,3H),5.20-5.26(m,2H),6.90(dd,J=5.2,7.3Hz,1H),7.00 (dd,J=1.6,7.3Hz,1H),7.18-7.22(m,2H),7.94-7.98(m,2H),8.19(dd,J= 1.6,5.2Hz,1H).
The minor isomer 4- { (1R) -1- [ (3S) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy-was also isolated]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } benzoic acid methyl ester, isomer 2(0.80g, 6%). LC/MS (m/z): 441.2(M + H).1H NMR(CDCl3)δ -0.06(s,9H),0.73-0.89(m,2H),1.50(d,J=7.2Hz,3H),1.51(s,3H), 3.23-3.33(m,2H),3.37(q,J=7.2Hz,1H),3.84(s,3H),4.87-4.93(m,2H), 6.88-6.92(m,2H),7.01(dd,J=5.3,7.3Hz,1H),7.58(dd,J=1.5,7.3Hz,1H), 7.73-7.76(m,2H),8.18(dd,J=1.5,5.3Hz,1H).
Preparation example 56
4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid methyl ester
Figure GDA0002319866120000521
To 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy ] at ambient temperature]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } benzoic acid methyl ester, isomer 1(10.16g,23.1mmol) in DCM (154mL) was added TFA (34.9mL, 461 mmol). After 17hr, the solution was concentrated under reduced pressure to give a residue. MeOH (308mL) and ethylenediamine (1.70mL,25.4mmol) were added to the residue. 5.0M aqueous NaOH was added slowly at RT until the mixture reached pH 10. After 2hr at RT, the solution was concentrated under reduced pressure to give a residue. DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM; the organic layers were then combined with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a crude product. The crude product was purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 4:1-0: 1). The impure chromatographic fractions were concentrated under reduced pressure and re-purified by flash chromatography on silica eluting with hexane/EtOAc (gradient 4:1-0: 1). All pure fractions from both chromatographic purifications were combined to give the title compound after evaporation of the solvent (5.11g, 71%). LC/MS (m/z): 311.0(M + H). Analysis of the material by chiral chromatography (column: Chiralpak AD-H4.6X 150 mm; eluent: 90: 10, EtOH: ACN (containing 0.2% IPAm); flow rate: 1.0mL/min, UV225nm) showed>99% ee, corresponding to the second eluting isomer, tR=5.42min。
Preparation example 57
4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid
Figure GDA0002319866120000522
To 4- { (1R) -1- [ (3) at ambient temperatureR) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b]Pyridin-3-yl]To a solution of methyl ethyl } benzoate (3.92g,12.6mmol) in THF (63.1mL) was added a solution of LiOH (907mg,37.9mmol) in water (12.6 mL). After 20hr, the reaction mixture was diluted with DCM and saturated aqueous NaCl solution. 1.0M HCl was added slowly at RT until the mixture reached pH 2. The layers were separated and washed with CHCl3The aqueous layer was extracted 3 times with 2-propanol (3: 1). The organic layers were combined and washed with Na2SO4Drying, filtration and concentration under reduced pressure gave the title compound (3.73g, 100%). LC/MS (m/z): 297.0(M + H).
Example 1: first method
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Figure GDA0002319866120000531
Scheme 3 step E: to 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] at RT]Pyridin-3-yl]To a solution of ethyl } pyridine-2-carboxylic acid (100mg,0.34mmol) and TEA (0.23 mL,1.68mmol) in DMF (2.0mL) was added 1- (2, 6-dimethylpyridin-4-yl) methylamine dihydrochloride (105mg,0.504mmol) and 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxatriphospha-cyclohexane-2, 4, 6-trioxide (50% by weight in EtOAc, 0.343mL,0.57 mmol). After 17hr, DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The residue was purified by flash chromatography on silica eluting with DCM/MeOH (gradient 1:0-9:1) to give the title compound (107mg, 75% yield) after evaporation of the solvent. LC-ES/MS (m/z): 416.2(M + H). The compound was purified by chiral SFC (column:
Figure GDA0002319866120000532
AD-H; eluent: 40:60, EtOH (containing 0.2% IPAm): CO2(ii) a Flow rate: 5mL/min, UV225nm) showed 97.4% ee, corresponding to the second washDe isomer, tR=2.95min.[α]D 20=-79.32 °(c=1.0,CHCl3)。
Example 1: second method
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
At RT to N- [ (2, 6-dimethylpyridin-4-yl) methyl]-5- { (1R) -1- [ (3R) -3-methyl-2-oxo-1- { [2- (trimethylsilyl) ethoxy]Methyl } -2, 3-dihydro-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl } pyridine-2-carboxamide (15.4g,28.2mmol) in DCM (308mL) was added in portions to TFA (154mL,2.04 mol). The reaction mixture was stirred at RT for 1.5hr, concentrated under reduced pressure, and dried in vacuo to give a residue. The residue was dissolved in THF (308mL) and NH was added in portions4OH (30%NH3154mL) resulting in a temperature rise to 45 ℃. The reaction mixture was cooled to RT, stirred for 40min and diluted with DCM and water. The layers were separated and the aqueous layer was extracted with DCM. The organic layers were combined and washed with saturated aqueous NaCl solution. The aqueous layer was re-extracted with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The residue was purified by flash chromatography on silica eluting with DCM/EtOAc/MeOH (gradient 50:50:0-47.5:47.5:5) to afford a solid. The solid was dried in a vacuum oven for 1 day. The solid was dissolved in MTBE (600mL) and EtOAc (100mL) and washed 2 times with water. The aqueous layer was extracted with EtOAc. The organic layers were combined and washed with Na2SO4Drying, filtering, and concentrating under reduced pressure to obtain solid. The solid was dried under a stream of nitrogen for 3 days and in a vacuum oven for 1 day. This material was dissolved in 50mL of 4:1EtOH: H2And O, concentrating under reduced pressure. The dissolution/concentration operation was repeated 2 times to obtain a solid. The solid was dried in a vacuum oven for about 20hr to give the title compound (5.9g, 50% yield). The compound was purified by chiral SFC (column:
Figure GDA0002319866120000541
AD-H; eluent: 40:60, EtOH (containing 0.2% IPAm): CO2(ii) a Flow rate: 5mL/min, UV225nm) analysis of the material showed 97.4%ee, corresponding to the second eluting isomer, tR=2.97min.LC-ES/MS(m/z):416.2(M+H)。
Example 1: third method
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Scheme 5 steps D and E: the following procedure can be carried out in two batches, the two batches being combined and then subjected to chromatography: reacting (3R) -3- [ (1R) -1- (6-bromo-3-pyridyl) ethyl]-3-methyl-1H-pyrrolo [2,3-b]Pyridin-2-one (72.4g,217.94mmol) was charged to a 2L Parr autoclave equipped with a mechanical stirrer containing dry toluene (925mL), phenol (22.7g,241.2mmol) and TEA (115.0g,1136.4 mmol). Sealing the autoclave with N2Fully purifying, adding Pd (OAc)2(500mg,2.2 mmol) followed by the addition of 4, 5-bis (diphenylphosphino) -9, 9-dimethylxanthene (1.3g,2.2 mmol). Again using N2The autoclave mixture was then purged with CO, pressurized to 60psi with CO, and heated to 85 deg.C overnight. The reaction mixture was cooled slightly, opened and (2, 6-dimethyl-4-pyridinyl) methylamine dihydrochloride (50.1g,239.6mmol) was added rapidly. The autoclave was sealed again and heated at 120 ℃ for 1 hr. The reaction mixture was cooled to RT, diluted with EtOAc (. about.1L), filtered through a bed of celite, and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with acetone in hexane (gradient 4:1-1:0) to give the crude title compound as a pale yellow solid after evaporation of the solvent. This material was dissolved in EtOAc (1L) and added
Figure GDA0002319866120000551
Metal Scavenger(200g,
Figure GDA0002319866120000552
) The resulting mixture was stirred at RT overnight. The mixture was filtered through a celite bed, washed with EtOAc (. about.2L), and the filtrate was concentrated under reduced pressure to give the title compound (181.3g, 77.2% yield, 2 runs combined) as a white solid. The reaction was performed by chiral FC (column:
Figure GDA0002319866120000553
AD-H; eluent: 40:60, EtOH (containing 0.2% IPAm): CO2(ii) a Flow rate: 5mL/min, UV225nm) analysis of the substance, tR2.95min, show>98%ee。LC-ES/MS(m/z):416.2(M+H).
Example 1A
Hydrated N- [ (2, 6-dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide hydrochloride
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide (8g,19.3mmol) was dissolved in 100mL of acetone at 1000rpm/60 ℃ to give a yellow solution. A1M HCl in EtOAc (40mL,40.08mmol) was added and a white gum precipitated out of solution quickly. The sample was stirred at 60 ℃ for 30min, at which time a slurry of white solid began to form in solution. After the complete slurry time, the sample was a thin layer of white solid under a light yellow solution. The white solid was isolated by vacuum filtration, dried on filter paper for 15min, and dried in a vacuum oven at 70 ℃ for 1hr to give the title product as a white crystalline solid in hydrated form (5.69g, 65.4% yield).
The crystalline hydrated HCl salt samples prepared were characterized by an XRD spectrum using CuKa radiation having diffraction peaks (2 theta values) as described in table 1 below, in particular having a peak at 20.7 ° and having one or more peaks selected from 19.8 °, 12.9 ° and 14.0 °; the error in diffraction angle was 0.2 degrees.
Table 1: x-ray powder diffraction Peak of hydrated N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide hydrochloride of example 1A
Peak(s) Angle (° 2 θ) +/-0.2 ° Relative intensity (% of the strongest peak)
1 12.9 47.1%
2 14.0 46.0%
3 16.1 30.8%
4 19.8 50.0%
5 20.7 100.0%
6 22.1 32.2%
7 23.3 23.8%
8 24.7 42.4%
9 26.0 16.1%
10 28.2 17.6%
Example 1B
Methanolic N- [ (2, 6-dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide hydrochloride
N- [ (2, 6-Dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide (366.0mg,0.88mmol) was dissolved in 2mL EtOAc at 1000rpm/60 ℃ to give a yellow solution. A1M HCl in EtOAc (1.25mL,1.84mmol) was added and a white solid precipitated out of solution immediately. The mixture was stirred into a slurry at 60 ℃ for 30min, at which time the solid was converted to a bright white particulate birefringent solid. The sample was cooled to 5 ℃ to give an off-white solid which was isolated by vacuum filtration. The solid cake was then rinsed with another 500 μ L of EtOAc, and the solid turned slightly brighter white. The cake was dried on filter paper for 15min and then dissolved in 1mL MeOH at 60 ℃. The sample was taken out and placed on a bench to cool, and then refrigerated at 5 ℃ for 2hr, and collected by vacuum filtration to obtain crystalline methoxide of HCl salt.
The crystalline methoxide sample of the HCl salt prepared was characterized by an XRD spectrum using CuKa rays having diffraction peaks (2 θ values) as described in table 2 below, in particular having a peak at 16.3 ° and having one or more peaks selected from 24.6 °, 20.8 ° and 20.1 °; the error in diffraction angle was 0.2 degrees.
Table 2: x-ray powder diffraction Peak of methanolized N- [ (2, 6-dimethylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-B ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide hydrochloride of example 1B
Peak(s) Angle (° 2 θ) +/-0.2 ° Relative intensity (% of the strongest peak)
1 12.5 8.7%
2 14.4 2.9%
3 15.5 2.4%
4 16.3 100.0%
5 20.1 14.1%
6 20.8 35.5%
7 22.4 4.6%
8 24.6 58.5%
9 25.6 3.9%
10 30.3 1.9%
Example 2
N- [ (2, 6-dimethyl-4-pyridyl) methyl ] -4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzamide
Figure GDA0002319866120000571
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ Me): to 4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2, 3-b)]Pyridin-3-yl]Ethyl radical]To a solution of benzoic acid (0.5g,1.503mmol) and (2, 6-dimethyl-4-pyridinyl) methylamine hydrochloride (426mg,3.0mmol) in DMF (4mL) were added TEA (628. mu.L, 3.0mmol) and T3P (50% by weight in EtOAc, 1.8mL,3.0mmol), and the resulting mixture was stirred at RT overnight. The reaction mixture was diluted with saturated aqueous NaCl and extracted with EtOAc. Separating the organic layer with Na2SO4Drying, filtration, concentration under reduced pressure, and purification by flash chromatography on silica gel, gradient eluting with 0-7% MeOH in DCM, gave the title compound after evaporation of the solvent (548mg, 88% yield). LC-ES/MS (m/z): 415.2(M + H).
Example 3
N- [ (2-Ethyl-6-methylpyridin-4-yl) methyl ] -4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzamide
Figure GDA0002319866120000581
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ Et): to a solution of 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (45mg,0.15mmol) in DMF (0.76mL) at RT was added 1- (2-ethyl-6-methylpyridin-4-yl) methylamine dihydrochloride (51mg,0.23mmol), DIPEA (0.15mL,0.91 mmol) and HATU (70mg,0.18 mmol). After 17hr, the reaction mixture was purified by reverse phase chromatography (Phenomenex Gemini-NX C18 column), eluting with 10mmol ammonium bicarbonate (pH 10 with 5% methanol) and ACN, and the solvent was evaporated to give the title compound (53.7mg, 83% yield). LC-ES/MS (m/z): 429.2(M + H).
Example 4
N- [ (2-cyclopropyl-6-methyl-4-pyridinyl) methyl ] -4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzamide
Figure GDA0002319866120000582
Scheme 9, step a (R ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ c-Pr): substantially by example 1: the procedure described in the first procedure was prepared from 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (50mg,0.17mmol) and (2-cyclopropyl-6-methyl-4-pyridinyl) methylamine dihydrochloride (48mg,0.2mmol) to give the title compound (73mg, 98% yield). LC-ES/MS (m/z): 441.2(M + H).
Example 5
N- [ (2-cyclobutyl-6-methylpyridin-4-yl) methyl ] -4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzamide
Figure GDA0002319866120000591
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ c-Bu): prepared essentially by the method described in example 3 from 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (45mg,0.15mmol) and (2-cyclobutyl-6-methyl-4-pyridinyl) methylamine dihydrochloride (57mg,0.23mmol) to give the title compound (58.5mg, 85% yield). LC-ES/MS (m/z): 455.2(M + H).
Example 6
N- [ (2-cyclopentyl-6-methylpyridin-4-yl) methyl ] -4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzamide
Figure GDA0002319866120000592
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ cyclopentyl): prepared essentially by the method described in example 3 from 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (45mg,0.15mmol) and (2-cyclopentyl-6-methyl-4-pyridinyl) methylamine dihydrochloride (60mg,0.23mmol) to give the title compound (65.1mg, 92% yield). LC-ES/MS (m/z): 469.2(M + H).
Example 7
N- [ (4, 6-Dimethylpyridin-2-yl) methyl ] -4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzamide
Figure GDA0002319866120000593
Scheme 9, step a (Q ═ N, X ═ CH, Y ═ N, Z ═ CH, R ═ Me): to 4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] at RT]Pyrimidin-5-yl]To a solution of ethyl } benzoic acid (300mg,1.01mmol) and TEA (0.422mL,3.03mmol) in DMF (6.0mL) was added 1- (4, 6-dimethylpyridin-2-yl) methylamine (Aldrich, CAS #76457-15-3, 206mg,1.51mmol) and 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxatriphospha-cyclohexane-2, 4, 6-trioxide (50% by weight in EtOAc, 1.03mL,1.72 mmol). After 3hr, DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers are combined and the organic layer is combined,with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The crude product was purified by flash chromatography on silica eluting with DCM/MeOH (gradient 1:0-9:1) to give the title compound (312mg, 74% yield) after evaporation of the solvent. LC-ES/MS (m/z): 416.2(M + H).
Example 8
N- [ (4, 6-Dimethylpyridin-2-yl) methyl ] -4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl } benzamide
Figure GDA0002319866120000601
Scheme 9, step a (Q ═ N, X ═ CH, Y ═ CH, Z ═ N, R ═ Me): to 4- { (1R) -1- [ (5R) -5-methyl-6-oxo-6, 7-dihydro-5H-pyrrolo [2,3-d ] at RT]Pyrimidin-5-yl]To a solution of ethyl } benzoic acid (316mg,1.06mmol) and TEA (0.44mL,3.19mmol) in DMF (6.3mL) was added 1- (2, 6-dimethylpyridin-4-yl) methylamine (188mg,1.38mmol) and 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxatriphospha-cyclohexane-2, 4, 6-trioxide (50% by weight in EtOAc, 1.08mL,1.81 mmol). After 4hr, DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The crude product was purified by flash chromatography on silica eluting with DCM/MeOH (gradient 1:0-9:1) to give the title compound (323mg, 73% yield) after evaporation of the solvent. LC-ES/MS (m/z): 416.2(M + H).
Example 9
N- [ (2-cyclopropyl-6-methylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Figure GDA0002319866120000611
Scheme 9, step a (Q ═ CH, X ═ N, Y ═ CH, Z ═ N, R-c-Pr): to 5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] at RT]Pyridin-3-yl]Ethyl pyridineTo a solution of-2-carboxylic acid (75mg,0.25mmol) and TEA (0.176mL,1.26mmol) in DMF (1.5 mL) was added 1- (2-cyclopropyl-6-methylpyridin-4-yl) methylamine dihydrochloride (89mg, 0.38mmol) and 2,4, 6-tripropyl-1, 3,5,2,4, 6-trioxatriphospha-cyclohexane-2, 4, 6-trioxide (50% by weight in EtOAc, 0.257mL,0.428 mmol). After 17hr, DCM and saturated NaHCO were added3Aqueous solution, and the layers were separated. The aqueous layer was extracted 2 times with DCM. The organic layers were combined and washed with Na2SO4Drying, filtering and concentrating under reduced pressure to obtain a residue. The crude product was purified by flash chromatography on silica eluting with DCM/MeOH (gradient 1:0-9:1) to give the title compound (75mg, 67%) after evaporation of the solvent. LC-ES/MS (m/z): 442.2(M + H).
Example 10
N- [ (2, 6-dimethyl-4-pyridinyl) methyl ] -5- [ (1R) -1- [ (5R) -5-methyl-6-oxo-7H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxamide
Figure GDA0002319866120000612
Scheme 9, step a (Q ═ N, X ═ N, Y ═ CH, Z ═ N, R ═ Me): prepared essentially by the method described in example 3 from 5- [ (1R) -1- [ (5R) -5-methyl-6-oxo-7H-pyrrolo [2,3-d ] pyrimidin-5-yl ] ethyl ] pyridine-2-carboxylic acid (100mg,0.334mmol) and (2, 6-dimethyl-4-pyridinyl) methylamine hydrochloride (1.5equiv.,0.51mmol) to give the title compound (87mg, 62% yield). LC-ES/MS (m/z): 417.2(M + H).
Example 11
N- [ (6-cyano-4-methyl-2-pyridyl) methyl ] -4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzamide
Figure GDA0002319866120000621
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ N, Z ═ CH, R ═ CN): substantially by example 1: the procedure described in the first method was prepared from 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (0.05g,0.17mmol) and 6- (aminomethyl) -4-methyl-pyridine-2-carbonitrile (0.03g,0.2mmol) to give the title compound (49mg, 68% yield). LC-ES/MS (m/z): 426.2(M + H).
Example 12
N- [ (4, 6-Dimethylpyridin-2-yl) methyl ] -4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3- ] pyridin-3-yl ] ethyl } benzamide
Figure GDA0002319866120000622
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ N, Z ═ CH, R ═ Me): prepared essentially by the method described in example 3 from 4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzoic acid (350mg,1.2mmol) and (4, 6-dimethyl-2-pyridinyl) methylamine (173mg,1.3mmol) to give the title compound (192mg, 39% yield). LC-ES/MS (m/z): 415.2(M + H).
Example 13
N- [ (2-Ethyl-6-methylpyridin-4-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Figure GDA0002319866120000623
Scheme 9, step a (Q ═ CH, X ═ N, Y ═ CH, Z ═ N, R ═ Et): prepared essentially by the method described in example 3 from 5- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] pyridine-2-carboxylic acid (45mg,0.15mmol) and (2-ethyl-6-methyl-4-pyridinyl) methylamine dihydrochloride (50mg,0.23mmol) to give the title compound (51.2mg, 79% yield). LC-ES/MS (m/z): 430.3(M + H).
Example 14
4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } -N- { [ 2-methyl-6- (propan-2-yl) pyridin-4-yl ] methyl } benzamide
Figure GDA0002319866120000631
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ i-Pr): prepared essentially by the method described in example 3 from 4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzoic acid (45mg,0.15mmol) and (2-isopropyl-6-methyl-4-pyridinyl) methylamine (37mg,0.23mmol) to give the title compound (57.5mg, 86% yield). LC-ES/MS (m/z): 443.3(M + H).
Example 15
N- [ (4, 6-Dimethylpyridin-2-yl) methyl ] -5- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } pyridine-2-carboxamide
Figure GDA0002319866120000632
Scheme 9, step a (Q ═ CH, X ═ N, Y ═ N, Z ═ CH, R ═ Me): prepared essentially by the method described in example 3 from 5- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] pyridine-2-carboxylic acid (45mg,0.15mmol) and (4, 6-dimethyl-2-pyridinyl) methylamine dihydrochloride (47mg,0.23mmol) to give the title compound (57.5mg, 86% yield). LC-ES/MS (m/z): 416.3(M + H).
Example 16
N- [ (2-cyano-6-methylpyridin-4-yl) methyl ] -4- { (1R) -1- [ (3R) -3-methyl-2-oxo-2, 3-dihydro-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl } benzamide
Figure GDA0002319866120000641
Scheme 9, step a (Q ═ CH, X ═ CH, Y ═ CH, Z ═ N, R ═ CN): prepared essentially by the method described in example 3 from 4- [ (1R) -1- [ (3R) -3-methyl-2-oxo-1H-pyrrolo [2,3-b ] pyridin-3-yl ] ethyl ] benzoic acid (45mg,0.15mmol) and 4- (aminomethyl) -6-methyl-pyridine-2-carbonitrile (70mg,0.18mmol) to give the title compound (50mg, 77% yield). LC-ES/MS (m/z): 426.3(M + H).
CGRP receptor antagonists inhibit cAMP production
hCGRP (human calcitonin gene-related peptide) receptor is functionally coupled to the G α s protein stimulation of hCGRP results in increased intracellular cAMP synthesis and can be blocked by the addition of a receptor antagonist.
Cell culture: (iii) cultured SK-N-MC neuroblastoma cells endogenously expressing hCGRP receptor (
Figure GDA0002319866120000642
HTB-10TM) After being supplemented with 10% heat-inactivated fetal bovine serum (FBS;
Figure GDA0002319866120000643
) Non-essential amino acids
Figure GDA0002319866120000644
Eagle's Minimum essential medium (HYCLONE) of 1mM sodium pyruvate, 2mM L-glutamine, 100U/mL penicillin, and 10. mu.g/mL streptomycinTM) Medium to about 70% confluence. After providing fresh medium, cells were incubated overnight at 37 ℃. On the day of measurement, use
Figure GDA0002319866120000645
(MPBiomedicals) cells were separated and resuspended in assay buffer [ Hank Balanced salt solution/Dulbecco phosphate buffered saline with 100mg/mL CaCl each mixed according to 1:22And MgCl23.3mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid, 0.03% bovine serum albumin and 0.5mM 1-methyl-3-isobutylxanthine (as inhibitor of cAMP)]3-5K/well were seeded into 384-well poly-D-lysine coated white plates (BD Biosciences).
Inhibition of cAMP production: for dose-response studies, compounds were serially diluted 1:3 in dimethylsulfoxide and then 1:10 in assay buffer. Human CGRP (0.8 nM; Bachem) as a receptor-specific agonist of the hCGRP receptor was mixed with the diluted compound,and as an aggressive stimulant with its EC80The concentration is added to the cells.
And (3) data analysis: the amount of intracellular cAMP was quantified using HTRF technology (Cisbio) according to the supplier's instructions. Briefly, cAMP-d2 conjugate and anti-cAMP-cryptate conjugate in lysis buffer were incubated with the treated cells for 90min at room temperature. Use of
Figure GDA0002319866120000651
The plate reader (Perkin-Elmer) immediately detects the HTRF signal to calculate the fluorescence ratio at 665-. Raw data were converted to cAMP amounts (pmole/well) using cAMP standard curves generated for each experiment. Using a four parameter logistic curve fitting program (
Figure GDA0002319866120000652
v5.3.1.22 or GENEDATA
Figure GDA0002319866120000653
v12.0.4) calculation of relative EC from the top-bottom range of the concentration-response curve50Value, K using the following equationbValue estimation as agonist-corrected IC50The value:
Kb=(IC50) /[1+ ([ agonist ]]/EC50)]。
Estimated KbValues are reported as mean ± SEM, averaged over the number of trials (n).
Following the procedure substantially as described above, the compounds of examples 1-16 have the K determined for the human CGRP receptor shown in Table 3b. This demonstrates that the compounds of examples 1-16 are antagonists of the human CGRP receptor in vitro.
TABLE 3 Kb determined in vitro on human CGRP receptor
Figure GDA0002319866120000654
Figure GDA0002319866120000661
CGRP (calcitonin gene-related peptide) non-human primate study
Capsaicin-induced skin blood flow (DBF) was used as a target to participate in biomarkers to assess CGRP receptor activity in non-human primates (NHPs). The method is based on an improvement of the earlier disclosed method [ Hershey et al, Regulatory peptides, Vol.127, pp.71-77,2005, 1-3 ].
Study population: animal studies can be performed under the protocol approved by the Covance institute of Animal Care and UseCommittee. Considering the close homology between NHPs and human CGRP receptors, cynomolgus (cynomolgus) NHPs may be used. The study population may include healthy, male cynomolgus NHPs that received CGRP antagonists for the first time, weighing 3-4 kg.
Cynomolgus NHPs were included in this study based on a pre-screen for capsaicin responsiveness. The study with the compound of example 1 included NHPs that showed > 50% increase in blood flow from baseline and stable physiological manifestations during imaging with 2mg (20 μ L/ring) topical capsaicin treatment (average of 3O-rings) as a response to capsaicin in the screening arm. NHPs were used for crossover design, where all NHPs received the full dose after a two-week washout period. Total n is 10 NHPs per group.
Dose administration: in Laser Doppler Imaging (LDI) experiments, NHPs received each oral administration of vehicle, 3, 10 and 30mg/kg of CGRP receptor antagonist example 1 (10% gum arabic w/v/0.05% antifoam 1510-US emulsion v/v/in pure water) 90 minutes prior to capsaicin administration.
Pharmacodynamic sampling: animals were fasted overnight before each capsaicin challenge. On the day of the experiment, NHPs were anesthetized with 1% isoflurane for about 30min and then scanned. The NHP was placed in a quiet temperature controlled room, supine on a warm mini surgical blanket, and the shaved arm was placed on a heating pad under the laser head. Three neoprene O-rings (size 8mm ID) were placed on the NHP forearm, approximately 1cm apart. During the 30 minute stabilization period, a preliminary scan was obtained to confirm the correct positioning of the O-ring. The baseline scan is acquired once the baseline temperature (about 37 ℃) stabilizes. After completion of the baseline scan, 20. mu.l capsaicin was addedSolution (in 170. mu.l EtOH, 80. mu.l)
Figure GDA0002319866120000662
20. 250 μ l of pure H250mg capsaicin in O solution) was applied to each O-ring. An additional 25min of continuous scans (85, 90, 95, 100, 105, 110, 115 and 120min after treatment with CGRP antagonist compound) were performed every 5 min.
Analysis and statistics: the LDI repeat scan was analyzed using Moor software v.5.3 (Moor instruments, Wilmington, DE) according to the signal analysis region of interest, and Microsoft Excel spreadsheets were used to average the signal from the region of interest at a given point in time. Changes in DBF are reported as percent changes from baseline DBF. [ two LDI data points were removed from the analysis due to statistically low exposure of the compound of example 1 (normalized residue method with 95% threshold)]. Inputting analytical data into Graphpad
Figure GDA0002319866120000671
4.0 drawing, use
Figure GDA0002319866120000672
The mixed effect model of repeated measures in 9.1 was statistically analyzed. Data are presented as mean +/-SEM.
Using a mixed effect model with repeated measurements (autoregressive correlation by AR1 procedure) and multiple adjustments of hair emergence rates, 3mg/kg, 10mg/kg and 30mg/kg of the compound of example 1 produced statistically significant reduced increases in blood flow after capsaicin challenge compared to vehicle with group mean inhibition of 32.1% (p <0.008), 46.1% (p <0.0004) and 58.8% (p <0.00002), respectively, providing evidence of CGRP target involvement.

Claims (23)

1. A compound of the formula:
Figure FDA0002287474000000011
wherein
Y is CH or N;
z is CH or N;
provided that when Y is CH, Z is N, and when Y is N, Z is CH;
x is CH or N;
q is CH or N; and is
R is C1-C3 alkyl, C3-C5 cycloalkyl or CN;
or a pharmaceutically acceptable salt thereof.
2. The compound or salt of claim 1, wherein Q is CH.
3. The compound of claim 1, or a salt thereof, wherein Y is CH and Z is N.
4. The compound of claim 2, or a salt thereof, wherein Y is CH and Z is N.
5. The compound or salt of any one of claims 1-4, wherein X is N.
6. The compound or salt of any one of claims 1-4, wherein R is C1-C3 alkyl.
7. The compound of claim 5, or a salt thereof, wherein R is C1-C3 alkyl.
8. The compound or salt of any one of claims 1-4, having the formula:
Figure FDA0002287474000000012
9. the compound of claim 5, or a salt thereof, having the formula:
Figure FDA0002287474000000021
10. the compound of claim 6, or a salt thereof, having the formula:
Figure FDA0002287474000000022
11. the compound of claim 7, or a salt thereof, having the formula:
Figure FDA0002287474000000023
12. the compound or salt of any one of claims 1-4, having the formula:
Figure FDA0002287474000000024
13. the compound or salt of claim 5, having the formula:
Figure FDA0002287474000000025
14. the compound or salt of claim 6, having the formula:
Figure FDA0002287474000000026
15. the compound or salt of claim 7, having the formula:
Figure FDA0002287474000000031
16. the compound or salt of claim 1, wherein the compound is:
Figure FDA0002287474000000032
17. the compound or salt of claim 16, wherein the compound is:
Figure FDA0002287474000000033
18. the compound or salt of claim 17, wherein the compound is:
Figure FDA0002287474000000034
19. the salt of claim 18, which is:
Figure FDA0002287474000000035
20. use of a compound or salt according to any one of claims 1-19 in the manufacture of a medicament for the treatment of migraine.
21. Use of a compound or salt according to any one of claims 1-19 in the manufacture of a medicament for the prevention of migraine.
22. A pharmaceutical composition comprising a compound or salt of any one of claims 1-19 and one or more pharmaceutically acceptable carriers, diluents, or excipients.
23. A process for preparing a pharmaceutical composition comprising admixing a compound or salt of any one of claims 1-19 and one or more pharmaceutically acceptable carriers, diluents, or excipients.
CN201680043902.0A 2015-08-12 2016-08-05 CGRP receptor antagonists Active CN107922406B (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201562203993P 2015-08-12 2015-08-12
US62/203,993 2015-08-12
US201562266769P 2015-12-14 2015-12-14
US62/266,769 2015-12-14
PCT/US2016/045693 WO2017027343A1 (en) 2015-08-12 2016-08-05 Cgrp receptor antagonists

Publications (2)

Publication Number Publication Date
CN107922406A CN107922406A (en) 2018-04-17
CN107922406B true CN107922406B (en) 2020-04-03

Family

ID=56852394

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201680043902.0A Active CN107922406B (en) 2015-08-12 2016-08-05 CGRP receptor antagonists

Country Status (7)

Country Link
US (1) US9637495B2 (en)
EP (1) EP3341370B1 (en)
CN (1) CN107922406B (en)
CA (1) CA2990739A1 (en)
ES (1) ES2879243T3 (en)
TW (1) TW201718574A (en)
WO (1) WO2017027343A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR111665A1 (en) 2017-05-15 2019-08-07 Lilly Co Eli DERIVATIVES OF 3-METHYL-PIRROLIDINE-2,5-DIONA USEFUL AS ANTAGONISTS OF THE CGRP RECEIVER
CN110678457B (en) 2017-05-15 2022-08-16 伊莱利利公司 CGRP receptor antagonists
CN111100128B (en) * 2018-10-26 2022-09-06 广安凯特制药有限公司 Synthetic method of Ribocini intermediate product and intermediate compound thereof
MA54823A (en) 2019-01-24 2022-04-27 Takeda Pharmaceuticals Co HETEROCYCLIC COMPOUND AND ITS USE
JP2021050161A (en) 2019-09-25 2021-04-01 武田薬品工業株式会社 Heterocyclic compound and use thereof
EP4073059A1 (en) 2019-12-12 2022-10-19 Eli Lilly and Company Cgrp antigonists useful as tracer compounds for positron emission tomography

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101421267A (en) * 2006-04-10 2009-04-29 默克公司 Process for the preparation of pyridine heterocycle CGRP antagonist intermediate
WO2010078307A1 (en) * 2008-12-29 2010-07-08 Xenon Pharmaceuticals Inc. Spiro-oxindole-derivatives as sodium channel blockers
CN102656159A (en) * 2009-10-14 2012-09-05 百时美施贵宝公司 Cgrp receptor antagonists
CN101448821B (en) * 2004-09-13 2013-03-27 默沙东公司 Carboxamide spirolactam cgrp receptor antagonists
WO2013066360A1 (en) * 2010-12-22 2013-05-10 Merck Sharp & Dohme Corp. Fused heterocyclic azaindane carboxamide cgrp receptor antagonists
WO2013169567A1 (en) * 2012-05-09 2013-11-14 Merck Sharp & Dohme Corp. Spirolactam cgrp receptor antagonists

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4960785A (en) 1988-12-16 1990-10-02 Pfizer Inc. Indolinone derivatives
AR074072A1 (en) 2008-11-11 2010-12-22 Lilly Co Eli COMPOSITE OF IMIDAZOL -PIPERIDIN -PIRROL-PIRIMIDIN-6-ONA, PHARMACEUTICAL COMPOSITION THAT INCLUDES IT AND ITS USE TO PREPARE A USEFUL MEDICINAL PRODUCT TO TREAT MULTIFORM GLIOBLASTOMA
WO2013169574A2 (en) * 2012-05-09 2013-11-14 Merck Sharp & Dohme Corp. Aliphatic spirolactam cgrp receptor antagonists

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101448821B (en) * 2004-09-13 2013-03-27 默沙东公司 Carboxamide spirolactam cgrp receptor antagonists
CN101421267A (en) * 2006-04-10 2009-04-29 默克公司 Process for the preparation of pyridine heterocycle CGRP antagonist intermediate
WO2010078307A1 (en) * 2008-12-29 2010-07-08 Xenon Pharmaceuticals Inc. Spiro-oxindole-derivatives as sodium channel blockers
CN102656159A (en) * 2009-10-14 2012-09-05 百时美施贵宝公司 Cgrp receptor antagonists
WO2013066360A1 (en) * 2010-12-22 2013-05-10 Merck Sharp & Dohme Corp. Fused heterocyclic azaindane carboxamide cgrp receptor antagonists
WO2013169567A1 (en) * 2012-05-09 2013-11-14 Merck Sharp & Dohme Corp. Spirolactam cgrp receptor antagonists

Also Published As

Publication number Publication date
US9637495B2 (en) 2017-05-02
ES2879243T3 (en) 2021-11-22
EP3341370B1 (en) 2021-04-28
EP3341370A1 (en) 2018-07-04
CA2990739A1 (en) 2017-02-16
TW201718574A (en) 2017-06-01
WO2017027343A1 (en) 2017-02-16
US20170044163A1 (en) 2017-02-16
CN107922406A (en) 2018-04-17

Similar Documents

Publication Publication Date Title
CN107922406B (en) CGRP receptor antagonists
CN107922385B (en) CGRP receptor antagonists
TW202043217A (en) Bcl-2 inhibitors
AU2016315360A1 (en) Pyridinone dicarboxamide for use as bromodomain inhibitors
CN111170938A (en) 5, 6-disubstituted pyridine-2-carboxamides as cannabinoid receptor agonists
CN110662741B (en) 3-methyl-pyrrolidine-2, 5-dione derivatives useful as CGRP receptor antagonists
WO2011143155A1 (en) Substituted n-heteroaryl tetrahydro-isoquinoline derivatives, preparation and therapeutic use thereof
ES2870303T3 (en) CGRP receptor antagonists
WO2021078120A1 (en) Compounds and compositions for the treatment of parasitic diseases
CN113121398B (en) Arylalkylamine compound, preparation method and medical application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20180705

Address after: indiana

Applicant after: Iran

Address before: indiana

Applicant before: Eli Lilly and Co.

GR01 Patent grant
GR01 Patent grant